WO2005118831A2 - Polymeric compositions and related methods of use - Google Patents

Polymeric compositions and related methods of use Download PDF

Info

Publication number
WO2005118831A2
WO2005118831A2 PCT/US2005/006418 US2005006418W WO2005118831A2 WO 2005118831 A2 WO2005118831 A2 WO 2005118831A2 US 2005006418 W US2005006418 W US 2005006418W WO 2005118831 A2 WO2005118831 A2 WO 2005118831A2
Authority
WO
WIPO (PCT)
Prior art keywords
dopa
mpeg
dhpd
peg
substrates
Prior art date
Application number
PCT/US2005/006418
Other languages
French (fr)
Other versions
WO2005118831A3 (en
Inventor
Philip B. Messersmith
Jeffrey Dalsin
Lijun Lin
Bruce P. Lee
Kui Huang
Original Assignee
Northwestern University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Northwestern University filed Critical Northwestern University
Priority to JP2007500804A priority Critical patent/JP5133048B2/en
Priority to AU2005250314A priority patent/AU2005250314A1/en
Priority to CA002557330A priority patent/CA2557330A1/en
Priority to EP05804747A priority patent/EP1735456A4/en
Publication of WO2005118831A2 publication Critical patent/WO2005118831A2/en
Publication of WO2005118831A3 publication Critical patent/WO2005118831A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/08Materials for coatings
    • A61L31/10Macromolecular materials
    • CCHEMISTRY; METALLURGY
    • C09DYES; PAINTS; POLISHES; NATURAL RESINS; ADHESIVES; COMPOSITIONS NOT OTHERWISE PROVIDED FOR; APPLICATIONS OF MATERIALS NOT OTHERWISE PROVIDED FOR
    • C09JADHESIVES; NON-MECHANICAL ASPECTS OF ADHESIVE PROCESSES IN GENERAL; ADHESIVE PROCESSES NOT PROVIDED FOR ELSEWHERE; USE OF MATERIALS AS ADHESIVES
    • C09J4/00Adhesives based on organic non-macromolecular compounds having at least one polymerisable carbon-to-carbon unsaturated bond ; adhesives, based on monomers of macromolecular compounds of groups C09J183/00 - C09J183/16

Definitions

  • MAPs Mussel adhesive proteins
  • DOPA L-3,4- dihydroxyphenylalanine
  • Control of cell and protein adhesion on surfaces is critical to the performance of biosensors, medical diagnostic products, any instrumentation and assays used requiring handling serum and other human/animal fluids, tissue engineering, localized in vivo drug delivery, implanted medical devices, healing of surgical incisions, adhesion of tissues such as bone and cartilage for healing, and nanotechnology (nanoparticle-based therapies and diagnostic tools).
  • control of cellular and protein adhesion to surfaces is also important. Such applications include prevention of mussel attachment to boats and ships, piers, and other structures used in oceans and fresh water, prevention of algal and bacterial growth on water lines used for industrial and drinking water, and sensors used to measure water quality and purity.
  • PAO poly(alkylene oxides)
  • PEG polyethylene glycol
  • PEO polyethylene oxide
  • PEO-PPO-PEO block copolymers such as those available under the PLURONICS brand name
  • PEG/tetraglyme poly(methoxyethyl methacrylate)
  • PMEMA poly(methoxyethyl methacrylate)
  • polyMPC Poly(methacryloyl phosphatidylcholine)
  • compositions which function e.g., as an adhesive, in a substantially aqueous environment.
  • the preferred compositions generally comprise an adhesive moiety and a polymer moiety, the polymer moiety having a desired surface active effect (or other desired characteristics).
  • the adhesive moiety of a composition of this invention comprises dihydroxyphenyl derivatives including, di (DHPD) wherein the second DHPD is
  • the polymer moiety comprises poly (alkyleneoxide).
  • the adhesive moiety comprises DHPD, e.g., DOPA (discussed herein), and the polymer moiety comprises PEO-PPO-PEO block polymers (also discussed above).
  • the adhesive moiety comprises DHPD including a pendent chain comprising ethylenic or vinylic unsaturation such as, for example, an alkyl acrylate.
  • this invention comprises dihydroxyphenyl (DHPD) adhesivie compound of formula (I) wherein
  • Ri and R 2 may be the same or different and are independently selected from the group consisting of hydrogen, saturated and unsaturated, branched and unbranched, substituted and unsubstituted C 1-4 hydrocarbon;
  • P is separately and independently selected from the group consisting of -NH 2 , - COOH, -OH, -SH,
  • Ri and R 2 are defined above. a single bond, halogen,
  • Ai and A 2 are separately and independently selected from the group consisting of H, a single bond; a protecting group, substantially poly(alkyleneoxide), D
  • R 4 R 4 is H, C
  • _ 6 lower alkyl, or poly(alkylene oxide)-C-C CH 2 ,
  • R 3 is defined as above, and D is indicated in Formula (i). 1]
  • the poly(alkylene oxide) has the structure
  • R 3 and R 4 are separately and independently H, or CH 3 and m has a value in the range between 1 and about 250, A 4 is NH 2 , COOH, OH, -SH, -H or a protecting group.
  • DHPD is
  • DHPD is of the structure:
  • a 2 is -OH and Ai is substantially poly(alkylene oxide) of the structure
  • R 3 , i and m being defined as in claim 2.
  • the poly(alklene oxide) is a block copolymer of ethylene oxide and propylene oxide.
  • a method of this invention involves adhering substrates to one another comprising the steps of providing DHPD of the structure:
  • Ri and R 2 are defined as above; applying the DHPD of the above structure to one or the other or both of the substrates to be adhered; contacting the substrates to be adhered with the DHPD of the above structure therebetween to adhere the substrates to each other, and optionally repositioning the substrates relative to each other by separating the substrates and recontacting them to each other with the DHPD of the above structure therebetween.
  • DHPD Dihydroxyphenyl derivatives
  • Ri and R 2 are defined below, and n ranges between 1 and about 5.
  • Ri and R 2 are hydrogen and P is, itself, dihydroxy phenyl.
  • a preferred DHPD in a practice of the present invention is 1-3,4, dihydroxy phenyl alanine (DOPA), (generically),
  • Subtantially poly(alkylene oxide) shall mean predominantly or mostly alkyloxide or alkyl ether in composition. This definition contemplates the presence of heteroatoms e.g., N, O, S, P, etc. and of functional groups e.g., -COOH, - NH 2 ,-SH, as well as ethylenic or vinylic unstaturation. It is to be understood any such non-alkyleneoxide structures will only be present in such relative abundance as not to materially reduce, for example, the overall surfactant, non-toxicity, or immune response characteristics, as appropriate, or of this polymer.
  • Figure 1 shows 1H NMR spectra of PLURONIC® F127, its carbonate intermediate (SC-PAO7) and DME-PAO7 in CDC1 3 .
  • Figure 2 provides differential scanning calorimetry thermograms of 30 wt
  • FIG. 3 plots shear storage modulus, G', of a 22 wt % DME-PAO7 aqueous solution as a function of temperature at 0.1 Hz and a strain of 0.45%. Shown in the inset is the rheological profile of a 22 wt % unmodified-PLURONIC® F127 aqueous solution as a function of temperature.
  • FIG. 4 plots shear storage modulus, G', of a 50 wt % DME-PAO8 aqueous solution as a function of temperature at 0.1 Hz and a strain of 0.45%. Shown in the inset is the rheological profile of a 50 wt % unmodified PLURONIC® F68 aqueous solution as a function of temperature.
  • Figure 5 plots storage moduli of DME-PAO8 aqueous solutions at 45 wt
  • Figures 6A and 6B show differential scanning calorimetry thermograms of (A) DOPA-PAO7 and (B) DME-PAO7 at different concentrations upon heating. Arrows indicate the location of gelation endotherm observed only at higher polymer concentrations.
  • Figures 7A-C show high-resolution C(ls) XPS peaks for (A) un-modified
  • Figures 8A-C provide TOF-SIMS positive spectrum showing peaks representing catechol binding of gold. Spectra were normalized to Au peak (m/z ⁇ 197).
  • Figure 9 provides TOF-SIMS spectra showing the positive secondary ion peak at mass m/z ⁇ 43 for unmodified Au substrate, Au exposed to mPEG-OH, mPEG- DOPA powder and Au exposed to mPEG-DOPA.
  • Figure 10 shows TOF-SIMS spectra showing the positive secondary ion peaks for Au substrate chemisorbed with mPEG-DOPA.
  • Catecholic binding of gold is observed at m/z ⁇ 225 (AuOC), 254 (AuOCCO), and 309. Less intense AuO a C b peaks are seen at m/z ⁇ 434, 450, 462, and 478.
  • Figure 11 shows SPR spectra of protein (0.1 mg/ml BSA) adsorption onto modified and unmodified gold surfaces.
  • mPEG-DOPA and mPEG-MAPd modified surfaces exhibited reduced protein adsorption compared to bare gold and mPEG-OH modified surfaces.
  • Figures 14 A-C are a series of SEM micrographs indicating the morphology of NIH 3T3 fibroblasts on (A) unmodified Au, (B) Au treated with mPEG-OH, and (C) mPEG-DOPA-modified Au. All treatments were at 50mg/ml in DCM for 24 h.
  • Figure 15 shows the UV/vis absorption spectrum of mPEG-DOPA stabilized magnetite nanoparticles suspended in several aqueous NaCl solutions at the concentrations as shown and plotted therein. Addition of NaCl did not induce nanoparticle precipitation.
  • Figure 16 shows addition of salt to untreated Au nanoparticles induces aggregation. Shown are UV/vis scans of lOnm untreated Au nanoparticles suspended in aqueous NaCl solutions (concentrations as shown and plotted therein). The attenuation and shift of the 520 nm absorption band with increasing NaCl concentration reflects aggregation of the nanoparticles.
  • Figure 17 illustrates addition of salt to mPEG-DOPA stabilized Au nanoparticles does not induce aggregation. Shown are UV/vis scans of lOnm mPEG- DOPA stabilized Au nanoparticles suspended in aqueous NaCl solutions (concentrations as shown and plotted therein). The lack of attenuation and shift of the 520nm absorption band with increasing NaCl concentration reflects effective stabilization of the nanoparticles.
  • Figure 18 plots the UV/vis absorption spectrum of mPEG-DOPA stabilized CdS nanoparticles suspended in aqueous NaCl solutions (concentrations as shown and plotted therein.
  • Figure 19 plots XPS survey scans of unmodified TiO 2 and TiO 2 treated with mPEG-DOPA ⁇ -3 .
  • Figure 20 plots the long-term resistance to cell adhesion on TiO 2 and TiO 2 modified with mPEG-DOPA ⁇ -3 .
  • the duration of the non-fouling response is proportional to the length of the DOPA peptide anchoring group. Adherent cells were visualized with calcium AM.
  • Figure 21 plots the high-resolution XPS scans of the Cis region of TiO 2 substrates modified with mPEG-DOPA 1-3 . Of note is the increase in the ether carbon peak (286. OeV) with increasing length of the DOPA peptide anchor.
  • Figure 22 plots the high-resolution XPS scans of the Ols region of TiO 2 substrates modified with mPEG-DOPA ⁇ -3 .
  • the peak at 532.9eV representing polymeric oxygen increases while the Ti-O-H peak (531.7eV) decreases with increasing DOPA peptide length.
  • Figure 23 plots the results of the Robust Design experiment on 316L stainless steel
  • Figure 24 plots the 4-hour cell attachment to a variety of surfaces modified by mPEG-DOPA ⁇ -3 using a 24-hour modification at 50°C at the indicated pHs.
  • Figure 25 plots the % gel conversion versus the UV exposure time in minutes.
  • Figure 26 plots the mole fraction of DOPA incorporated versus the mol % of 1 or 7 in the precursor solution.
  • Figure 27 plots the % gel conversion versus mol % 1 or in the precursor solution.
  • Figure 28 X-ray Photoelectron Spectroscopy XPS analysis of a silicon nitride surface.
  • Figure 29 is a free monitoring of functionalized silicon nitride cantilevers.
  • Figure 30 is an analysis of entropic elasticity of poly(ethylene glycol).
  • Figure 31 is a force measure of side chain modified DOPA.
  • Figure 32 is a proposed model of DOPA-T 1 O 2 binding mechanism.
  • Figure 33 is an atomic force microscopy arrangement.
  • Figure 34 is data regarding force measurement.
  • Figure 35 is adhesion data.
  • Figure 36 is synthetic route and data analysis.
  • DHPD dihydroxyphenyl derivative
  • DHPD adhesives and polymeric compositions have many uses, including prevention of protein and/or cell adhesion to a surface in various medical, industrial and consumer applications.
  • the DHPD adhesives can also be used as substitutes for sutures for a wound and as aids in healing bone fractures or cartilage-to-bone damage. These and other uses will be described in more detail below.
  • compositions of the present invention have the following structure:
  • Pi and P 2 are separately and independently defined as P in formula (I);. n and m independently range from 0 to about 5, provided that at least one of n or m is at least 1;
  • Adhesive Moiety is a dihydroxyphenyl derivative (“DHPD”) having the following preferred structure:
  • the DHPD adhesive can function in an aqueous environment.
  • an aqueous environment is any medium comprising water. This includes without limitation water, including salt water and fresh water, cell and bacterial growth media solutions, aqueous buffers, other water-based solutions, and body fluids.
  • the DHPD moiety can be derivatized. As would be understood by those skilled in the art, such derivatization is limited by the retention of the desired adhesive characteristic.
  • polymeric components providing a surface active effect and other desired characteristics will be well-known to those skilled in the art made aware of this invention.
  • the desired surface active effect relates to reduced particulate agglomeration and anti-biofouling, including resistance to cell and/or protein adhesion.
  • the polymer component can be water soluble, depending upon end-use application, and/or capable of micelle formation depending upon various other end-use applications.
  • Polymers useful in the present invention include, but are not limited to, polyethylene glycol (PEG), polyethylene oxide (PEO), polypropylene oxide (PPO), PEO-PPO-PEO block copolymers, polyphenylene oxide, PEG/tetraglyme, PMEMA, polyMPC, and perfluorunated polyethers.
  • the polymeric compositions can be synthesized in several ways.
  • the polymeric compositions may be synthesized through a general synthetic procedure for polymer end-group activation.
  • Various polymers or monomeric components thereof can be activated using carbonate chemistry.
  • a succinimidyl carbonate- activated polymeric component reacted with DHPD moiety can provide a stable urethane conjugate.
  • Two of the many possible pathways (a) and (b) in Scheme la and lb, below, show coupling with a poly(alkylene oxide) in either aqueous or non-aqueous solvents, without compromising desired bioadhesion.
  • a DHPD residue can be coupled to a polymeric component to provide the desired conjugate composition, through either urethane or amide bond formation.
  • a carboxylic acid group of the DHPD component can be esterified or derivatized with various other functional groups.
  • the DHPD component can be coupled to a polymeric component (e.g., amidation or esterification depending on polymer end group, -NH 2 or -OH) providing a DHPD functionality which can be derivatized by any of numerous known protecting groups, including without limitation Boc, Fmoc, borate, phosphate, and tributyldimethylsilyl.
  • the present invention is also a method of using urethane synthesis to incorporate a DHPD residue into a polymeric system.
  • a method includes (1) providing a polymeric component terminating in a plurality of monomers, each having a functional end group; (2) preparing a carbonate derivative of the polymeric component; and (3) preparing a urethane moiety upon reaction of the carbonate derivative and at least one DHPD moiety.
  • a polymeric component utilized in conjunction with this method can include those having terminal monomeric functionality reactive with a reagent providing the desired carbonate derivative and, ultimately, providing a urethane moiety coupling the polymeric and DHPD components.
  • a reagent providing the desired carbonate derivative and, ultimately, providing a urethane moiety coupling the polymeric and DHPD components.
  • Various other coupling reagents and/or hydroxy-terminating polymeric components can be used to provide the desired urethane moiety.
  • the present invention is also a method of using a carbonate intermediate to maintain catecholic functionality of a DHPD-incorporated polymeric composition and/or system, or to otherwise enhance the adhesion properties thereof.
  • a method includes (1) providing a polymeric component terminating in a plurality of monomers each having a functional end group; (2) reacting the polymeric component with a reagent to provide a carbonate intermediate; and (3) reacting the carbonate intermediate with at least one DHPD moiety.
  • this inventive method can be considered a way of enhancing the reactivity of the polymeric component end group, via a suitable carbonate intermediate. Subsequent reaction at the amino-nitrogen of DHPD moiety provides the corresponding conjugate while maintaining catecholic functionality.
  • DOPA methyl ester prepared by the reaction of DOPA with methanol in the presence of thionyl chloride, can be used in organic solvents. Reaction progress can be monitored by TLC and NMR, with the coupling reaction virtually complete in one hour (with representative conjugates DME-PAO7 (from PAO PLURONIC® F127) and DME-PAO8 (from PAO PLURONIC® F68)). High product yields were obtained upon purification from cold methanol.
  • the free carboxylic form of DOPA can be coupled with the carbonate intermediate in alkaline aqueous solution. It is well known that the chief difficulty in working with DOPA is its ease of oxidation (to DOPA-quinone and other products), which readily occurs in alkaline aqueous solutions.
  • a borate-protected DOPA can be first formed by adding DOPA to aqueous sodium borate (Scheme lb). The resulting complex is remarkably stable in neutral or alkaline solutions, and can be readily deprotected under acidic conditions.
  • DOPA was coupled to the ends of several commercially-available PAOs under alkaline aqueous conditions to yield DOPA-PAO7 and DOPA-PAO8.
  • Visual inspection of the reaction solution revealed the absence of strongly absorbing DOPA-quinone, an indication that DOPA remains unoxidized during the reaction.
  • acidification with HCI resulted in deprotection of the DOPA endgroups of the block copolymer.
  • the product yields (shown in Table 1) of selected DOPA-modified PAOs synthesized in aqueous solution were found to be lower than those synthesized in organic solvent. This may be due to the surfactant properties of the starting PAO material, causing the low efficiency of extraction of DOPA-modified PAO with dichloromethane from water. It should be noted that the free carboxylic acid in DOPA-PAO7 and DOPA-PAO8 can be further functionalized using standard peptide chemistry to tailor the properties of the block copolymers.
  • the four DOPA-modified PAOs of Table 1 could be stored at -20°C indefinitely with no discoloration or change in properties.
  • Control of cell and protein adhesion on surfaces is critical to the performance of biosensors, medical diagnostic products, any instrumentation and assays used requiring handling serum and other human/animal fluids, tissue engineering, localized in vivo drug delivery, implanted medical devices, healing of surgical incisions, adhesion of tissues such as bone and cartilage for healing, and nanotechnology (nanoparticle-based therapies and diagnostic tools).
  • the polymeric compositions of the present invention can be used as coatings to prevent protein and cellular adhesion to devices for medical and research applications. These include without limitation such uses as coatings for medical implants, coatings for surgical devices, coatings for devices that handle serum and other animal or human derived materials, medical diagnostic devices, and biosensors.
  • the polymeric compositions can be tissue adhesive polymeric hydrogels for medical uses such as tissue sealants, gels for prevention of surgical adhesion (scar tissue formation), bone and cartilage adhesives, tissue engineering, and site specific drug elution and for research uses such as immobilization of proteins including antibodies and small molecule analytes including pharmaceuticals.
  • tissue adhesive polymeric hydrogels for medical uses such as tissue sealants, gels for prevention of surgical adhesion (scar tissue formation), bone and cartilage adhesives, tissue engineering, and site specific drug elution and for research uses such as immobilization of proteins including antibodies and small molecule analytes including pharmaceuticals.
  • these coatings and hydrogels including without limitation prevention of marine biofouling (attachment of algae, bacteria, and mussels to surfaces underwater), prevention of bacteria contamination of water streams to industrial plants such as electronic and drug manufacturers, prevention of bacterial contamination of drinking water streams, dental and denture adhesives, underwater adhesives to deliver indicators, coatings for water purity and measurement sensors, paints used for prevention of biofouling, and use in cosmetics to adhere desired fragrances and colorants to hair, eyelids, lips, and skin, to form temporarily skin coloring such as tattoos and the like, and for resealable adhesives for consumer products such as storage bags.
  • Adhesive hydrogels can be also formed using the present methods.
  • the DHPD adhesive is attached to polymers capable of forming a hydrogels in vivo or in vitro.
  • hydrogels can be formed by a number of methods including the use of self-assembling polymers that form gels at higher temperatures such as normal human body temperatures, the use of polymers that can be cross-linked by an enzymatic reaction, the use of polymers that can be subjected to oxidation to form cross-linked hydrogels, and the use of polymers that can be subjected to photoactivation to produce cross-linked hydrogels.
  • the anti-biofouling coatings of the present invention may be applied to medical devices, such as vascular or arterial stents, pacemakers, heart valves, glucose monitors and other biosensors, vascular wraps, defibrillators, orthopedics devices, and surgical devices, including sutures and catheters.
  • medical devices such as vascular or arterial stents, pacemakers, heart valves, glucose monitors and other biosensors, vascular wraps, defibrillators, orthopedics devices, and surgical devices, including sutures and catheters.
  • the polymeric compositions of the present invention can be used as coatings to prevent protein and/or cellular adhesion to a device for medical and research applications. These include without limitation such uses as coatings for medical implants, coatings for surgical devices, coatings for devices that handle serum and other animal or human derived materials, medical diagnostic devices, and biosensor.
  • a surface may be modified by the polymeric composition of the present invention in any number of ways.
  • the polymeric composition may be absorbed onto the surface or a DHPD moiety containing a polymerization initiator may be adsorbed onto the surface and polymer growth initiated from the surface.
  • a number of polymerization techniques are possible, including without limitation surface initiated radical polymerization, radical polymerization methods, ionic polymerization, ring-opening polymerization, and photopolymerization.
  • the surface density of polymer can be increased by treating surfaces with PEG solutions near the lower critical solution temperature (LCST), or cloud point.
  • LCST critical solution temperature
  • This approach is useful for polymers that show inverse solubility transitions at high temperature and high ionic strength, such as poly(ethylene glycol), poly(N-isopropylacrylamide) and other N-substituted poly(acrylamides) that show inverse solubility transitions.
  • DHPD moieties in the adhesive component and the pH of the modification buffer are responsible for most of the variation in the cell and/or protein adhesion resistance of the modified materials.
  • adsorption time and polymeric composition concentration contributes little to the variation in cell and/or protein adhesion resistance of the modified materials.
  • the greater the number of DHPD moiety monomers in the adhesive component the better the cell and/or protein adhesion resistance.
  • the density of the polymeric composition on the surface correlates well with resistance to cell and/or protein adhesion.
  • the thickness of the coating layer can be from about 20 A to about 100 ⁇ m, including 30 A, depending on the polymer composition used and the pH of the modification buffer.
  • the concentration of the polymer composition used for modification of a surface can be from about .1 mg/ml to about 75 mg/ml.
  • the pH of the modification buffer can be from about 3 to about 9.
  • the modification time can be from about 10 minutes to about 72 hours.
  • the temperature of the modification can be from about 25°C to about 60°C.
  • XPS survey scans of unmodified TiO 2 revealed strong peaks at -458 eV (Ti2p) and -530 eV (Ols) characteristic of native oxide, as well as a small peak at 248.7 eV (Cis) as a result of adventitious hydrocarbon contamination.
  • TiO 2 substrates treated with mPEG-DOPA 1-3 under cloud point conditions demonstrated dramatic increases in surface-bound carbon as reflected by the Cis peak, suggesting the presence of PEG on the surface.
  • the increases in the Cis peaks observed after modification with mPEG-DOPA ⁇ -3 were directly proportional to the number of terminal DOP As present.
  • a small peak at 400 eV ( ⁇ ls) was seen in the spectrum of the TiO 2 surface modified with mPEG-DOPA ⁇ -3 , representing the amide nitrogen in DOPA.
  • Table 2 shows the titanium, oxygen, and carbon atomic composition calculations for TiO 2 - modified with mPEG-DOPA ⁇ -3 .
  • the oxygen signal is further subdivided into metal oxide (Ti-O-Ti), surface hydroxide (Ti-O-H), and organic oxygen and coupled water (C- O, H 2 O) species.
  • Table 2 Titanium, oxygen, and carbon atomic composition calculations
  • DOPA creates strong, reversible bonds with TiO 2 .
  • the energy of the bond is 30.56 kcal/mol and needs about 800 pN to be detached from TiO 2 at the single molecule level, which is four times stronger than the interaction between Avidin and Biotin.
  • the DOPA-TiO 2 strength of interaction is about midway between that of Avidin-Biotin, one of the strongest hydrogen bond based interactions in biology (0.1-0.2 nN) and a covalent bond (>2nN).
  • DOPA conjugated cantilevers displayed significant adhesion accompanied by entropic elasticity of the PEG chain (Fig 34).
  • a histogram of the force distribution shows a uni-modal shape indicating only single adhesive event, which is different compared to the case of a multivalent protein, Avidinl2.
  • the length of stretched PEG (36 nm) was consistent with the expected contour length of a PEG molecule (37 nm, Fig 30).
  • d is the z-displaced distance of piezoelectric device when a single DOPA-PEG molecule was fully stretched during retraction (Fig 34C).
  • the 'd' values appeared to be almost constant throughout many repeated cycles although it did vary slightly (Fig 34A). This small variation might be due to DOPA binding to the surface at different angles.
  • An important feature of our experiment is that the unbinding signals are epetitions using the 'identical' DOPA molecule. This is compared to the traditional approach of single molecule pulling experiments where tips picked up one molecule randomly. This also demonstrates that the DOPA adhesion chemistry was completely reversible.
  • Mussels developed an interesting way to create such a strong binding in water, a post-translational modification of tyrosine by tyrosine hydroxylase.
  • This enzyme catalyzes a reaction of adding one hydroxyl group using tyrosine as a substrate and a large amount is found in threads and plaques where DOPA exists as well. It is surprising to say that the small post-translational modification (-OH) seems to produce a huge change of adhesive ability. Thus, experiments were designed to show a correlation between the posttranslational modification and binding ability.
  • a tyrosine tethered cantilever was prepared instead of DOPA, and tyrosine adhesion on TiO2 was investigated. No detectable force signals were observed except some non-specific adhesions with low probability (Fig 35A).
  • the TiO2 surface was replaced with gold.
  • the aromatic ring of tyrosine binds to a gold surface in a parallel orientation to the surface through ⁇ - ⁇ electron interaction, which is a well-known mechanism in surface adsorption chemistryl8,19.
  • the same cantilever used in TiO2 produced relatively strong adhesions repeatedly on gold surfaces (Fig 35B).
  • DOPA Biological roles of DOPA go beyond adhesiveness upon oxidation: it crosslinks polypeptide chains resulting in stiffer materials found throughout threads and pads.
  • the crosslinking mechanism has multiple pathways starting from a chemically unstable DOPAquinone structure.
  • Aryl-aryl ring coupling (di-DOPA) has been found in mussel adhesive proteins 20 but Michael addition (quinone-alkylamine adducts) products have been found in other species not mussels (Fig 36A). Therefore, these structures may occur as results of oxidation in mussels as well. It is clear in terms of crosslinking but is under debate with respect to adhesive properties after maturation i.e. oxidation. It was demonstrated that the DOPAquinone structure is not a major player for adhesiveness.
  • DOPAquinone-PEG chain is spatially and chemically stabilized by excess co- conjugation of methoxy-PEG molecules (5-10 molar equivalent) which is an important molecular configuration for preventing further reactions of DOPAquinone.
  • measured AFM signals showed two clear distributions in terms of force magnitude: high force and low force (Fig 36B).
  • Statistical analysis of the data yielded two clear histograms with 178 ⁇ 62 pN for low and 741 ⁇ 110 pN for high force (Fig 36C).
  • the quinone binding can be assigned to the low force region because it appeared only after the oxidation was triggered and subsequently became more frequent over time (Fig 36D).
  • the slow kinetic feature of DOPA oxidation contributed to initial high frequency of DOPA signals. This is the first single molecule experiment about detecting the structural change of a small molecule upon external stimulus. Based on these results, the possibility of the DOPAquinone structure being responsible for the high adhesiveness can be ruled out. Therefore, without being bound to any theory, the regeneration of reduced form i.e. di-hydroxyl group of DOPA during oxidation, is believed to be a very important requirement for maintaining or changing adhesive properties of DOPA containing materials at an interface.
  • the DOPA anchoring system can be a new platform to study other extensible biological macromolecules such as polysaccharides, DNA, and proteins.
  • This method is also highly contrasted with the conventional single molecule experiments where a tip 'sees' different molecules at every single movement of a cantilever. It has been a big barrier to investigate molecular responses upon external stimuli if a given stimulus was not hundred percent effective23,.
  • the DOPA-based anchorage system can be an alternative technique to overcome these problems in current single molecule pulling experiments.
  • a picture describes how the blue mussel (Myt ⁇ lus Edulis) sticks to metal oxide surfaces.
  • the circle included one plaque where the unusual amino acid, DOPA, was found.
  • (B) Two major protein components uncovered in plaques in mussels, Mefp-3 and Mefp-5. These mussel adhesive proteins have high content of DOPA: 27 mole % of Mefp-5 and 21 % of Mefp-3.
  • (C) AFM tip modifications Polymerization of 3- aminopropyltrimethoxysilane (APTMS) introduced amine groups on Si3N4 tip surfaces (not drawn). A long chain describes a PEG molecule conjugated with single (Boc)-DOPA at the end. Mixture of mPEG-NHS(2k) and Fmoc-PEG-NHS(3.4k) at a molar ratio of 5-10:1 was used to stabilize DOPA-PEG molecule (see supplemental section for details).
  • APIMS 3- aminopropyltrimethoxysilane
  • (D) A plot of bonding strength (linear) vs. loading rate (log). Loading rate was the product of spring constant of a cantilever and a pulling speed. Four different loading rates were selected: 1500, 180.7, 28.4 and 2 nN/sec. Averaged forces with standard deviation were plotted at each given loading rate. Forces were 846.48 ⁇ 157 pN (1500 nN/s), 781 ⁇ 151 pN (180 nN/s), 744 ⁇ 206 pN (28.4 nN/s), and 636.2 ⁇ 150 (2 nN/s). [0099] (E) A schematic energy landscape of DOPA binding.
  • DOPA was created by the action of tyrosine hydrxylase and subsequently oxidized to DOPAquinone by pH and the enzyme. It is unstable and reactive due to tendency of radical formation of DOPAquinone. It can crosslink with other DOPA molecules (di- DOPA) as well as reacts with amine groups from lysines. The arrows are the potential reactions found in other species not in mussel adhesive proteins.
  • DOPA signals (circle, left y-axis) gradually decreased from twenty-two events for the first ten minutes to only three events during the last time window.
  • quinone signals (triangle, right y-axis) increased from one event at the first time window to forty-two events at the last time window (50-60 min).
  • the antifouling coating of the present invention can either be essentially permanent i.e., lasting 120 days or more, or biodegradeable depending on the number of DOPA or DOPA-derived moieties in the adhesive component.
  • Figure 20 shows the results of a 28-day 3T3 fibroblast cell adhesion and spreading assay on TiO 2 treated with mPEG-DOPA 1-3 . At early time points (i.e.
  • the protein and cell attachment resistance correlates well with the length of the DOPA peptide anchoring group, with resistance increasing in the order mPEG-DOPA ⁇ mPEG-DOPA 2 ⁇ mPEG- DOPA 3 .
  • TiO 2 substrates treated with mPEG-DOPA 2 and mPEG-DOPA 3 maintain reduced cell attachment, or resistance, through 21 days.
  • Robust design methodology was used to determine the effect of DOPA peptide anchor length and modification conditions (pH, concentration and time) on the PEG surface density and antifouling performance of metal, metal oxide, semiconductor, and polymer surfaces. The nine experiments utilized for each substrate are described below in Table 7.
  • the polymeric compositions of the present invention can be also used to coat the surfaces of devices and instrumentation used for handling body fluids including sera.
  • the coating on the surface of the device or instrument blocks protein binding to the surfaces thus reducing or eliminating the need for extensive washing or cleaning of the device or instrument between uses.
  • the devices need to be thoroughly cleaned to prevent cross contamination between samples of bodily fluids applied top the device.
  • caustic agents such as 50% bleach and/or elevated temperatures.
  • the coating process would be to circulate an aqueous solution of 1 mg/ml of DHPD polymer through the device at room temperature for a period of a few hours.
  • the coatings of the present invention can be used on medical implants for a wide variety of uses.
  • the coatings can be used to block bacterial adhesion and therefore growth on the implanted device reducing the possibility of infection at the site of implant.
  • the coatings can be used to reduce the amount of acute inflammation on the device by reducing protein binding and cell adhesion to the device.
  • the coatings of the present invention can also be used as nanoparticles to prevent aggregation of these particle in the presence of serum. Hydrogels
  • the polymeric compositions of the present invention can also be as surgical adhesives for medical and dental uses and as vehicles for drug delivery to mucosal surfaces.
  • the polymeric compositions can be used as tissue adhesive polymeric hydrogels for medical uses such as tissue sealants, gels for prevention of surgical adhesions (scar tissue formation), bone and cartilage adhesives, tissue engineering, and site specific drug elution and for research uses such as immobilization of proteins including antibodies and small molecule analytes including pharmaceuticals.
  • the polymeric compositions of the present invention may also be used as interfacial bonding agents, wherein the neat monomers or solution of monomers are applied to a surface as a primer or bonding agent between a tissue surface or a metal or metal oxide implant/device surface and a bulk polymer or polymer hydro gel.
  • the polymeric compositions of the present invention can be injected or delivered in a fluid form and harden in situ to form a gel network.
  • the in situ hardening can occur through photocuring, chemical oxidation, enzymatic reaction or through the natural increase in temperature resulting from delivery into the body.
  • the present invention is also a method for the non-oxidative gelation of a polymeric composition of the present invention.
  • One such method includes (1) providing a polymeric composition of the present invention; (2) admixing water and the polymeric composition; and (3) increasing admixture temperature sufficient to gel the polymeric composition, such temperature increase without oxidation of the polymer or DOPA or DOPA-derived moiety residue incorporated therein.
  • an increase in admixture concentration can reduce the temperature required to effect gelation.
  • a larger hydrophilic block thereof can increase the temperature required to gel the corresponding composition.
  • Various other structural and/or physical parameters can be modified to tailor gelation, such modifications as can be extended to other polymeric compositions and/or systems which are consistent with the broader aspects of this invention.
  • PLURONIC® block copolymers self-assemble in a concentration- and temperature-dependent manner into micelles consisting of a hydrophobic PPO core and a water-swollen corona consisting of PEO segments.
  • certain PEO-PPO-PEO block copolymers such as PLURONIC® F127 and PLURONIC® F68, transform from a low viscosity solution to a clear thermoreversible gel at elevated temperature. While not wanting to be bound by theory, it is generally assumed that the interactions between micelles at elevated temperature lead to the formation of a gel phase, which is stabilized by micelle entanglements.
  • micellization and gelation processes depend on factors such as block copolymer molecular weight, relative block sizes, solvent composition, polymer concentration, and temperature. For example, increasing the length of the hydrophilic PEO blocks relative to the hydrophobic PPO block results in an increase in micellization and gelation temperature (E ge ⁇ ).
  • DSC Differential scanning calorimetry
  • PLURONIC® F127, DME-PAO7 and DOPA-PAO7 were found to be qualitatively similar and were characterized by a large endothermic transition corresponding to micelle formation followed by a small endotherm at T gs ⁇ ( Figure 2).
  • the transition temperature of the small peak was found to correlate strongly with ge ⁇ determined by rheometry and the vial inversion method (Table 4).
  • Table 4 Gel temperatures obtained from vial inversion method, rheology or differential scanning calorimetry for 22 wt % DME-PAO7, DOPA-PAO7 and PLURONIC® F127 solutions.
  • DOPA-PAO7 copolymers and 35 to 54 % (w/w) of DOPA-PAO8 copolymers were prepared by the cold method, in which DOPA conjugate was dissolved in distilled water at about 4°C with intermittent agitation until a clear solution was obtained. Thermal gelation of concentrated solutions was initially assessed using the vial inversion method. In this method, the temperature at which the solution no longer flows is taken as the gelation temperature.
  • the gelation temperature was found to be strongly dependent on copolymer concentration and block copolymer composition (i.e., PAO7 versus PAO8).
  • PAO7 block copolymer composition
  • 22 wt % solutions of DOPA-P AO7 and DME-PAO8 were found to form a transparent gel at approximately 22.0 ⁇ 1.0°C; decreasing the polymer concentration to 18 wt % resulted in a gelation temperature of approximately 31.0 ⁇ 1.0°C.
  • DOPA-PAO7 solutions with concentrations less than 17 wt % did not form gels when heated to 60°C.
  • DOPA-PAO7 exhibits a slightly higher gel temperature than that (17.0 ⁇ 1.0°C) of unmodified PLURONIC® F127.
  • DOPA- PAO8 The gelation behavior of DOPA- PAO8 was found to be qualitatively similar, except that much higher polymer concentrations were required to form a gel. 54 wt % solutions of DOPA-PAO8 and DME-PAO8 formed gels at 23.0 ⁇ 1.0°C, while 50 wt % of DOPA-PAO8 gels at 33.0 ⁇ 1.0°C. However, DOPA-PAO8 solutions with concentrations less than 35 wt % did not form gels when heated to 60°C. DOPA-PAO8 exhibits a much higher gel temperature than that (16.0 ⁇ 1.0°C) of unmodified PLURONIC® F68. These gels were found to be resistant to flow over long periods of time.
  • FIG. 4 Shown in Figure 4 are the rheological profiles of 50 wt % solutions of unmodified PLURONIC® F68 and DME-PAO8 as a function of temperature.
  • the E ge ⁇ of a 50 wt % DME-PAO8 solution was found to be 34.1 ⁇ 0.6°e, whereas the r ge ⁇ of an equivalent concentration of unmodified PLURONIC® F68 was approximately 18°C lower (16.2 ⁇ 0.8°C).
  • the plateau storage moduli of 50 wt % solutions of DME-PAO8 and unmodified PLURONIC® F68 were not significantly different, approaching a plateau value as high as 50 kPa.
  • T ge ⁇ The concentration dependence of T ge ⁇ is illustrated in Figure 5, which shows the rheological profile of DME-PAO8 at two different concentrations as a function of temperature. Jgei of 45 wt % solution of DME-PAO8 was observed to be approximately 12°C higher than that of 50 wt % solution of DME-PAO8.
  • both DOPA and DOPA methyl ester can be considered hydrophilic, the increase of r ge ⁇ observed in the DOPA-modified PLURONIC® PAOs, compared with that of unmodified PLURONIC® PAOs, is likely due to the increase in length of the hydrophilic PEO segments resulting from coupling of DOPA to the endgroups.
  • micellization peak was seen to extend to temperatures above the onset of gelation, indicating that additional monomers aggregate into micelles at temperatures above the gelation point.
  • concentration dependence of DOPA-PAO7 and DME- PAO7 aggregation is shown in Figure 6.
  • DSC thermograms indicate a decrease in micellization temperature and ge ⁇ with increasing polymer concentration.
  • the broad endothermic peak corresponding to micellization can also be observed in solutions at concentrations at which no gelation takes place; the characteristic temperature of the broad peak increases linearly with decreasing copolymer concentration, whereas the small peak was observed to coincide to the gel temperature of the concentrate copolymers but disappears as copolymer concentration decreases.
  • various polymeric compositions of this invention can be designed and prepared to provide various micellization and/or thermal gelation properties.
  • degradation into excretable polymer components and metabolites can be achieved using, for instance, polyethylene glycol and lactic/glycolic acids, respectively.
  • the polymeric compositions of this invention provide improved adhesion by incorporation of one or more DHPD residues, such incorporation resulting from the coupling of a terminal monomer of the polymeric component to such a residue.
  • a photocurable DHPD-moiety-containing monomer is copolymerized with PEG-DA (PEG-diacrylate) to form adhesive hydrogels through photopolymerization.
  • Photopolymerization can be achieved at any visible of UV wavelength depending on the monomer used. This is decidedly determined by one skilled in the art.
  • the photocurable monomers consist of an adhesive moiety coupled to a polymerizable monomer with a vinyl group, such as a methacrylate group with or without an oligomeric ethylene oxide linker or fluorinated ether linker in between.
  • a photoinitiator such as 2,2'-dimethoxy-2-phneyl-acetonephenone (DMPA), camphorquinone/4-
  • gel conversion as determined by measuring the mass of the sondgel reached more than 75 wt % after 2 minutes of UV irradiation and increased to greater than 85 wt % upon irradiation for more than 5 minutes.
  • Gelation of PEG-DA occurred in 4 minutes or less when visible light initiators were used (4 minutes for CQ/DMAB and 3 minutes for AA/FNa 2 ).
  • Copolymerization of PEG-DA with 1 or 7 was qualitatively similar to polymerization of pure PEG-DA, although addition of 1 or 7 to the PEG-DA precursor solution resulted in a decrease in gel conversion that was dependent on DOPA monomer concentration and initiating system.
  • gel conversion was reduced to less than 85 wt % in the presence of 2.5 mol % or more of 1 or 7.
  • the extent of gel conversion was not statistically different between the gels. Similar DOPA concentration dependent inhibition was observed for the visible light induced initiators.
  • Figure 26 shows the mole fraction of DOPA incorporated into the gel network, as a function of mol % monomer 1 and 7 in the precursor solution. There was no significant difference in the mole fraction of DOPA incorporated between samples containing 1 and 7. As much as 24.9 ⁇ mol/g of DOPA was incorporated into the PEG hydrogels.
  • Direct evidence for the presence of DOPA in the gels was obtained by immersing the intact dialyzed hydrogels in nitrite reagent followed by NaOH. The initially colorless gels turned bright yellow after the addition of the nitrite reagent and then red following the addition of excess base. This color transition is typical of catechols, indicating that the unoxidized form of DOPA was incorporated into the hydrogels through photopolymerization. The intensity of the red color also reflects the concentration of DOPA incorporated into the photocured gels.
  • Equation (1) where R is the radius of curvature of the hemispherical gel. Load versus displacement data was fitted with Equation (1), which allowed the elastic moduli to be calculated based on the proportionality factor of the curve fit. As seen in Table 6, average Young's moduli (E) for DOP A-containing gel of around 50 kPa was obtained. Table 6 Average Young's Moduli for DOPA containing gels
  • modulus values are about 30% lower than that of PEG-DA gels, confirming the inhibitory effect of DOPA on radical photopolymerization.
  • DOP A-containing gels still exhibited moduli suitable for many biomedical applications.
  • a suitable modulus is one greater than 500 Pa.
  • Another use of the adhesive hydrogels is for localized drug delivery.
  • adhesive hydrogels can be formed on a mucous membrane in the mouth or oral cavity.
  • the hydrogels can be loaded with a drug such as an antibiotic and facilitate slow release of the drug over a period of time. They hydrogel can also be loaded with an analgesic and used to deliver pain relief at a localized site.
  • hydrogel can also be loaded with a chemotherapy drug and inserted into malignant tissue to deliver localized cancer therapy.
  • the hydrogel can also be loaded with a cell proliferation inhibitor therapeutic drug and used as a coating stent or other vascular device and used to control cell proliferation at the site of an implant of the vascular device.
  • a tissue adhesive hydrogel capable of being cross-linked in vivo can be used as a tissue sealant for replacing metal or plastic sutures.
  • the adhesive bends to the surrounding tissue at a surgical or injury site and the polymer forms a cohesive link to close the wound.
  • the hydrogel can also be used for repair of bone fractures and cartilage to bone damage.
  • compositions of this invention can include but are not limited to a urethane moiety between each such terminal monomer and DOPA residue.
  • a moiety is a synthetic artifact of the agent/reagent utilized to couple the DOPA residue with the polymeric component.
  • various other moieties are contemplated, as would be understood by those skilled in the art made aware of this invention, depending upon terminal monomer functionality and choice of coupling agent.
  • L-DOPA thionyl chloride, methacroyloyl chloride, /-butyldimethylsilyl chloride (TBDMS-C1), di-t-butyl dicarbonate, methacrylic anhydride, 2,2'-dimethoxy-2-phenyl-acetonephenone (DMPA), acryloyl chloride, 1,8- diazabicyclo[5.4.0]undec-7-ene (DBU), tetrabutylammonium fluoride (TBAF), 4- (dimethylamino)-benzoic acid (DMAB), l-vinyl-2 -pyrrolidone (VP), N,N-disuccinimidyl carbonate, sodium borate, sodium molybdate dihydrate, sodium nitrite, 4- (dimethylamino)pyridine (DMAP), ⁇ -hydroxysuccinimide, ⁇ , ⁇ -diisopropylethylamine, dimethylformamide, and dichlorome
  • Camphorquinone (CQ) was obtained from Polysciences, Inc. (Warrington, PA). Acetone was dried over 4A molecular sieve and distilled over P 2 O 5 prior to use. Triethylamine was freshly distilled prior to use. All other chemical reagents were used as received.
  • L-DOPA methyl ester hydrochloride was prepared according to the procedure of Patel and Price, J. Org. Chem., 1965, 30, 3575, which is incorporated herein by reference.
  • Shearwater Polymers, Inc. Hauntsville, AL
  • Ethyl acetate saturated with HCl was prepared by bubbling HCl gas through ethyl acetate (50 mL) for approximately 10 minutes.
  • Glass coverslips (12mm dia.) used in the following examples were cleaned by immersing in 5% Contrad70 solution, a detergent which is an emulsion of anionic and nonionic surfactants in an allealtine aqueous base (Decon Labs, Inc., Bryn Mawr, PA) in an ultrasonic bath for 20 minutes, rinsed with deionized (DI) H 2 O, sonicated in DI H 2 O for 20 minutes, rinsed in acetone, sonicated in acetone for 20 minutes, rinsed in hexanes, sonicated in hexanes for 20 minutes, rinsed in acetone, sonicated in acetone for 20 minutes, rinsed in DI H 2 O, and sonicated in DI H 2 O for 20 minutes.
  • DI deionized
  • coverslips were subsequently air-dried in a HEPA-filtered laminar flow hood.
  • clean coverslips were sputtered (Cressington 208HR) with 2 nm Cr followed by 10 mn Au (99.9% pure).
  • Titanium oxide (TiO 2 ) surfaces were prepared by electron beam physical evaporation onto silicon (Si) wafer and cleaned in a plasma chamber prior to testing.
  • Si wafers MEMC Electronic Materials, St. Peters, MO, surface orientation (100)
  • the Si wafer was then cut into 8mm x 8mm pieces which were subsequently cleaned by ultrasonication in the following media: 5% Contrad70, ultrapure water (ultrapure water is deionized and distilled), acetone, and petroleum ether.
  • the substrates were further cleaned in an oxygen plasma chamber (Harrick Scientific, Ossining, NY) at ⁇ 200 mTorr and 100W for 3 minutes.
  • XPS X-ray photoelectron spectroscopy
  • BIACORE 2000 (Biacore International AB; Uppsala, Sweden) using bare gold sensor cartridges.
  • the resonance response was calibrated using 0-lOOmg/ml NaCl solutions.
  • Dilute solutions (0.1 mM in H 2 O) of mPEG-DOPA, mPEG-MAPd, and mPEG-OH were injected into the SPR flow cell for 10 minutes after which flow was switched back to pure DI H O.
  • BSA bovine serum albumin
  • NIH 3T3-Swiss albino fibroblasts obtained from ATCC (Manassas, VA) were maintained at 37°C and 10% CO in Dulbecco's modified Eagle's medium (DMEM; Cellgro, Herndon, VA) containing 10% (v/v) fetal bovine serum (FBS) and lOOU/ml of both penicillin and streptomycin.
  • DMEM Dulbecco's modified Eagle's medium
  • FBS fetal bovine serum
  • lOOU/ml fetal bovine serum
  • modified and unmodified substrates were pretreated in 12-well TCPS plates with 1.0 ml of DMEM containing 10% FBS for 30 minutes at 37°C and 10% CO 2 .
  • Fibroblasts of passage 12-16 were harvested using 0.25% trypsin-EDTA, resuspended in DMEM with 10% FBS, and counted using a hemocytometer.
  • Cells were seeded at a density of 2.9 x 10 3 cell/cm 2 by diluting the suspension to the appropriate volume and adding 1 ml to each well.
  • the substrates were maintained in DMEM with 10% FBS at 37°C and 10% CO 2 for 4 hours, after which time unattached cells were aspirated.
  • Adherent cells on the substrates were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently treated with 5 ⁇ M l,l'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil; Molecular Probes, Eugene, OR) in DMSO for 30 minutes at 37°C.
  • the stain was then aspirated and substrates were washed (3x) with DMSO for 10 minutes and mounted on glass slides using Cytoseal (Stephens Scientific, Kalamazoo, MI) to preserve fluorescence.
  • PLURONIC® F127 (0.60 mmols) was dissolved in 30 mL of dry dioxane.
  • N,N J -Disuccinimidyl carbonate (6.0 mmols) in 10 mL dry acetone was added.
  • DMAP (6.0 mmols) was dissolved in 10 mL dry acetone and added slowly under magnetic stirring. Activation proceeded 6 hours at room temperature, after which SC-PAO7 was precipitated into ether. The disappearance of the starting materials during the reaction was followed by TLC in chi oroform-m ethanol (5:1) solvent system. The product was purified by dissolution in acetone and precipitation with ether, four times. The product yield was 65%.
  • PAO7 conjugate was used to prepare and purify DOPA-PAO8.
  • the product yield was
  • PLURONICs® F127 and F68 were determined using the colorimetric method of Waite and Benedict. Briefly, samples were analyzed in triplicate by diluting aliquots of standards or unknown solutions with 1 N HCl to a final volume of 0.9 mL. 0.9 mL of nitrite reagent (1.45 M sodium nitrite and 0.41 M sodium molybdate dihydrate) was added to the DOPA solution, followed immediately by the addition of 1.2 mL of 1 N NaOH. Due to time-dependent changes in absorbance intensity, care was taken to ensure that the time between the addition of NaOH and recording of the absorbance was 3 minutes for all standards and samples. The absorbance was recorded at 500 nm for all standards and samples. DOPA was used as the standard for both the DOPA methyl ester and DOPA conjugates. Example 8 Rheology
  • Bohlin VOR Rheometer Bohlin Rheologi, Cranbury, NJ. A 30 mm diameter stainless steel cone and plate geometry with a cone angle of 2.5 degrees was used for all measurements. The temperature was controlled by a circulating water bath. Samples were cooled in the refrigerator prior to transfer of 0.5 mL of liquid solution to the apparatus. Measurements of storage and loss moduli, G' and G", were taken in the oscillatory mode at 0.1 Hz and a strain of 0.45%. The heating rate was 0.5°C/min except in the vicinity of the gelation temperature, when it was reduced to 0.1°C/min. The strain amplitude dependence of the viscoelastic data was checked for several samples, and measurements were only performed in the linear range where moduli were independent of strain amplitude. Mineral oil was applied to a ring surrounding the outer surfaces of the sample compartment to prevent dehydration during measurements.
  • DSC Differential Scanning Calorimetry
  • the crude product was concentrated under reduced pressure and purified by column chromatography on Sephadex® LH-20 with methanol as the mobile phase.
  • the product, mPEG-DOPA was further purified by precipitation in cold methanol three times, dried in vacuum at room temperature, and stored under nitrogen at -20°C.
  • DHPD adhesive component Regardless of a particular DHPD adhesive component, a variety of polymeric components can be used in accordance with the synthetic techniques and procedures described above.
  • the polymeric component can vary in molecular weight limited only by corresponding solubility concerns.
  • a variety of other polymers can be used for surface anti-fouling and/or particle stabilization, such polymers including but not limited to hyaluronic acid, dextrans and the like.
  • the polymeric component can be branched, hyperbranched or dendrimeric, such components available either commercially or by well-known synthetic techniques.
  • Example 10a is the amidation product of the referenced starting materials
  • comparable polymer-DHPD conjugates can be prepared coupling the N-terminus of a DHPD component to an end group, back bone or side chain of a suitably functionalized natural or synthetic polymer, including those described above.
  • a suitable polymeric component terminating with a carbonate functionality can be used to provide the desired conjugate by reaction with the N-terminus of the desired DHPD component.
  • Example 11a The consensus decapeptide repeat sequence (mussel adhesive protein decapeptide, MAPd, NH 2 -Ala-Lys-Pro-Ser-Tyr-Hyp-Thr-DOPA-Lys-CO 2 H) of the blue mussel Mytilus edulis foot protein 1 (Mefp 1) was synthesized by solid phase peptide synthesis on Rink resin (0.6 mMol/g) using Fmoc protected amino acids, BOP, HOBt, and DIEA as activating agents, and NMP as solvent. Fmoc deprotection was performed using a 25% piperidine solution in NMP for twenty minutes.
  • the PEG-decapeptide conjugates (mPEG-MAPd, 2k or 5k) were cleaved at 0°C for two hours using 1 M TMSBr in TFA, with EDT, thioanisole, and m-cresol.
  • the crude mPEG-MAPd products were precipitated in ether at 0°C, and purified by preparative HPLC using a Vydac 218TP reverse phase column (220x 22mm x lO ⁇ m). The purity of the products was determined to be >90% using analytical HPLC, and the structures confirmed using a PerSeptive Biosystem MALDI-TOF-MS.
  • Example lib [00163] The synthesis and procedures of Example 11a can be extended analogous to and consistent with the variations illustrated in Example 10b.
  • other conjugates can be prepared using DOP A-containing polymers prepared by enzymatic conversion of tyrosine residues therein.
  • Other techniques well-known in the field of peptide synthesis can be used with good effect to provide other desired protein sequences, peptide conjugates and resulting adhesive/anti-fouling effects.
  • Example 12a [00164] Gold surfaces were modified by adsorption of mPEG-DOPA or mPEG-
  • MAPd (2k, 5k) from solution in DCM or phosphate-buffered saline (PBS; pH 3, 7.4, and 11) at polymer concentrations ranging from 0.1-75 mg/ml.
  • Time-of-flight SIMS data corroborated the XPS findings.
  • TOF-SIMS analysis was carried out on unmodified and mPEG-DOPA-modified Au substrates, as well as mPEG-DOPA powder and a gold substrate exposed to mPEG-OH. Data was collected from each substrate for about 4 minutes.
  • Example 12c [00173]
  • the modification illustrated in Example 12a can be extended to other noble metals, including without limitation, silver and platinum surfaces.
  • Such application can also be extended, as described herein, to include surface modification of any bulk metal or metal alloy having a passivating or oxide surface.
  • bulk metal oxide and related ceramic surfaces can be modified, as described herein.
  • Example 13 Silicate glass surfaces (glass coverslips) were modified by adsorption of mPEG-MAPd (2k) from a 10 mM solution in water, using the method described in Example 12a. The cell density of NIH 3T3 cells attached to modified and unmodified glass surfaces were evaluated as described, above.
  • mPEG-DOPA 50 mg was dissolved in water (18 M ⁇ -cm, Millipore) and combined with 1 mg of magnetite (Fe 3 O 4 ) powder. Similar preparations were also prepared using a mPEG-NH 2 (5k) (Fluka) and a mPEG-OH (2k) (Sigma) as controls. Each of these aqueous solutions was sonicated using a Branson Ultrasonics 450 Probe Sonicator for one hour while being immersed in a 25°C bath. The probe had a frequency of 20 kHz, length of 160 mm, and tip diameter of 4.5 mm.
  • Example 14b mPEG-DOPA stabilized nanoparticles were characterized by transmission electron microscopy (TEM), thermogravimetric analysis (TGA), fourier transform infrared spectroscopy (FTIR), and UV/vis spectroscopy. TEM results demonstrated that the majority of nanoparticles were of diameter of 5-20 nm (data not shown). TGA analysis of 0.4 mg of mPEG-DOPA stabilized magnetite indicated that the particles contain 17% by weight mPEG-DOPA (data not shown).
  • Example 14c The dry PEG-DOPA stabilized magnetite nanoparticles readily dispersed in aqueous and polar organic solvents (e.g., dichloromethane) to yield clear brown suspensions that were stable for months without the formation of noticeable precipitates.
  • aqueous and polar organic solvents e.g., dichloromethane
  • Suspensions of mPEG-DOPA stabilized nanoparticles in various solvents were prepared by dispersing 1 mg of mPEG-DOPA treated magnetite in 1 ml of water (18 M ⁇ -cm filtered using a Millex® AP 0.22 ⁇ m filter (Millipore)), DCM or Toluene. Suspensions were placed in a bath sonicator for ten minutes to disperse the nanoparticles. All three solutions were stable at room temperature for at least six months, whereas control suspensions of unmodified magnetite and magnetite stabilized by mPEG-OH or mPEG- NH 2 precipitated out in less than 24 hours in each solvent.
  • Example 14d Suspensions of mPEG-DOPA stabilized nanoparticles were also found to be stable under physiologic concentrations of salt.
  • mPEG-DOPA treated magnetite was placed in a quartz cuvette and combined with 0.7 ml of water (18 M ⁇ -cm filtered using a 0.25 ⁇ filter).
  • Aliquots of saturated NaCl solution (5 ⁇ l,10 ⁇ l, 20 ⁇ l, 50 ⁇ l, 100 ⁇ l) were sequentially added to the cuvette and allowed to stand for ten minutes before UV-VIS spectra were taken (Figure 15).
  • Example 15a Demonstrating stabilization of metal nanoparticles, commercial gold colloid suspension (Sigma, particle size 5 or 10 nm) was placed inside dialysis tubing (M w cutoff of 8000 for 5nm and 15000 for 10 nm) and dialyzed in ultrapure water for 2-3 days to remove the sodium azide present in the commercial preparation. The dialyzed suspensions were then placed into small glass vials and mPEG-DOPA added (10 mg/ml).
  • Example 15b [00181] Various other metal nanoparticles, including but not limited to, silver, platinum and the like can be stabilized as described in the preceding example. While stabilization was demonstrated using a representative conjugate composition of this invention, various other compositions can be prepared analogous to and consistent with the alternate embodiments described in Examples 10b and l ib.
  • Example 16a The data of this example demonstrates stabilization of semiconductor nanoparticles.
  • CdS nanoparticles Quantum dots
  • Fresh stock solutions (2 mM) of Cd(NO 3 ) 2 and Na 2 S were prepared in nanopure water.
  • the Na 2 S solution was injected slowly into 50 ml of Cd(NO 3 ) 2 solution using a gastight syringe at a rate of 20 ⁇ l s "1 .
  • the solution turned yellow with the addition of Na 2 S, and after 2 mL of Na 2 S was injected, a yellow precipitate appeared due to the aggregation of CdS nanoparticles.
  • the CdS precipitate was isolated and dried for further use. Using the method described above for magnetite, the dry CdS powder was dispersed in a mPEG-DOPA solution by sonication to yield a clear yellow solution. The yellow aqueous suspension was stored in the dark for several months at room temperature without visible formation of precipitate. Control experiments performed in the absence of polymer and in the presence of mPEG-OH or mPEG-NH 2 yielded yellow precipitate and a clear, colorless supernatant.
  • Example 16b [00183] The results of this example illustrate the in situ formation of stabilized semiconductor nanoparticles.
  • CdS nanoparticles (quantum dots) were formed in the presence of mPEG-DOPA by slowly mixing dilute methanolic solutions of Cd(NO 3 ) 2 and Na 2 S. Freshly prepared stock solutions (2 mM) of Cd(NO 3 ) 2 and Na 2 S were prepared in methanol.
  • Example 16c The polymeric conjugate compositions of this invention can also be used to stabilize a variety of other semiconductor materials. For instance, core-shell nanoparticles can be surface stabilized in accordance herewith.
  • Example 17 The optimization experiments of Examples 17-20 were performed with mPEG-DOPA-5K. Several parameters were examined to optimize the adsorption of mPEG-DOPA onto gold from solution, including type and pH of solvent, time of adsorption, and mPEG-DOPA solution concentration.
  • Example 20 [00188] The morphology of fibroblasts cultured on both unmodified and PEG- modified surfaces was examined via electron microscopy (Hitachi 3500 SEM). Fibroblasts on unmodified Au and mPEG-OH-modified Au were generally flat and well spread, while those cultured on mPEG-DOPA modified Au were far less spread ( Figures 14A-C).
  • FIG. 13 illustrates the differences in attachment and spreading of fibroblasts on bare Au, mPEG-OH-treated Au, and Au modified with mPEG-DOPA 5K, mPEG-MAPd 2K, or mPEG-MAPd 5K under optimal conditions (50 mg/ml for 24 hours).
  • the surfaces modified with DOP A-containing conjugates have significantly less cellular adhesion and spreading than either of the other two surfaces.
  • the mPEG-MAP 5K modification though, accounted for a 97% reduction in total projected cellular area and a 91% reduction in density of cells on the surface, a far greater reduction than that achieved by mPEG-DOPA 2K.
  • N-hydroxysuccinimide (0.110 g, 0.95 mmol) was added to a solution of Boc-DOPA(TBDMS) 2 (0.500 g, 0.95 mmol) in dry dichloromethane (DCM) (8.0 mL). The solution was stirred on an ice bath, and 1,3-dicyclohexylcarbodiimide (DCC) (0.197 g, 0.95 mmol) was added under nitrogen atmosphere. The reaction was stirred for 20 minutes at 0°C and then warmed to room temperature and stirred for an additional 4 hours. The reaction mixture was filtered to remove the urea byproduct and subsequently evaporated to 1/5 of its original volume.
  • DCM dry dichloromethane
  • Boc-DOPA(TBDMS) 2 -OSu (0.567 g, 0.91 mmol) was dissolved in dry dimethylformamide (DMF) (2.5 mL), and DOPA(TBDMS) 2 (0.405 g, 0.95 mmol) was added at once under a nitrogen atmosphere. The mixture was stirred on an ice bath, and diisopropylethylamine (DIEA) (158 ⁇ L, 0,91 mmol) was added dropwise via a syringe.
  • DIEA diisopropylethylamine
  • Example 21 The procedure of Example 21 was repeated using Boc-DOPA 2 (TBDMS) to obtain Boc-DOPA 2 (TBDMS) 4 -OSu.
  • Example 24 Synthesis ofBoc-DOPA 3 (TBDMS) 6
  • Example 22 The procedure of Example 22 was repeated using Boc-DOPA 2 (TBDMS) 4 -
  • Boc-DOPA 2 (TBDMS) 4 (0.5 g, 0.54 mmol) was dissolved in saturated
  • Boc-DOPA 3 (TBDMS) 6 (1.06 g, 0.79 mmol) was dissolved in saturated
  • Solid metal substrates Al, 316L stainless steel and NiTi
  • Si wafers were ground and polished, ultimately with 0.04 m colloidial silica (Syton, DuPont).
  • Si wafers were evaporated with either 20 nm TiO 2 or 10 nm TiO 2 /40 nm Au using an Edwards FL400 electron beam evaporator at ⁇ 10 "6 Torr and were subsequently diced in 8 mm x 8 mm pieces. All substrates were cleaned ultrasonically for 20 minutes in each of the following: 5% Contrad70 (Fisher Scientific), ultrapure H 2 O, acetone, and petroleum ether.
  • TiO 2 substrates were modified under cloud point conditions by immersion in mPEG-DOPA 1-3 solutions in 0.6 M K 2 SO 4 buffered with 0.1 M N- morpholinopropanesulfonic acid (MOPS) at 50°C for 24 hours. Modified substrates were rinsed with ultrapure H 2 O and dried under a stream of nitrogen.
  • MOPS N- morpholinopropanesulfonic acid
  • 316L Stainless Steel (Goodfellow, Devon PA) was modified under cloud point conditions by immersion in mPEG-DOPA ⁇ -3 solutions in 0.6 M K 2 SO 4 buffered with 0.1 M N-morpholinopropanesulfonic acid (MOPS) at 50°C for 24 hours. Modified substrates were rinsed with ultrapure H 2 O and dried under a stream of nitrogen.
  • MOPS N-morpholinopropanesulfonic acid
  • Al 2 O 3 (Goodfellow, Devon PA) was modified under cloud point conditions by immersion in mPEG-DOPA 1-3 solutions in 0.6 M K 2 SO 4 buffered with 0.1 M N-morpholinopropanesulfonic acid (MOPS) at 50°C for 24 hours. Modified substrates were rinsed with ultrapure H 2 O and dried under a stream of nitrogen.
  • MOPS N-morpholinopropanesulfonic acid
  • SiO 2 (1500A thermal oxide, University Wafer, South Boston, MA) was modified under cloud point conditions by immersion in mPEG-DOPA 1-3 solutions in 0.6 M K 2 SO buffered with 0.1 M N-morpholinopropanesulfonic acid (MOPS) at 50°C for 24 hours. Modified substrates were rinsed with ultrapure H 2 O and dried under a stream of nitrogen.
  • MOPS N-morpholinopropanesulfonic acid
  • NiTi alloy (10mm x 10mm x 1mm) was obtained from Nitinol Devices &
  • Au electro beam evaporated onto Si Wafer from University Wafer
  • mPEG-DOPA 1-3 solutions in 0.6 M K 2 SO buffered with 0.1 M N-morpholinopropanesulfonic acid (MOPS) at 50°C for 24 hours.
  • Modified substrates were rinsed with ultrapure H 2 O and dried under a stream of nitrogen.
  • Au 2 O 3 (Au samples as described in Example 28f were exposed to an oxygen plasma to form Au2O3) was modified under cloud point conditions by immersion in mPEG-DOPA ⁇ -3 solutions in 0.6 M K 2 SO buffered with 0.1 M N- morpholinopropanesulfonic acid (MOPS) at 50°C for 24 hours. Modified substrates were rinsed with ultrapure H 2 O and dried under a stream of nitrogen.
  • MOPS N- morpholinopropanesulfonic acid
  • GaAs Universal Wafer, South Boston, MA
  • MOPS N-morpholinopropanesulfonic acid
  • 3T3 Swiss albino fibroblasts (ATCC, Manassas, VA) of passage 12-16 were cultured normally at 37°C and 5% CO 2 in Dulbecco's modified Eagle's medium (DMEM) (Cellgro, Herndon, VA) supplemented with 10% fetal bovine serum (FBS) (Cellgro, Herndon, VA), 100 g/mL penicillin, and 100 U/mL steptomycin. Prior to cell adhesion assays, fibroblasts were harvested using 0.25% trypsin-EDTA, resuspended in growth medium, and counted with a hermacytometer.
  • DMEM Dulbecco's modified Eagle's medium
  • FBS fetal bovine serum
  • Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO 2 . Cells were seeded onto the substrates at a density of 2.9 x 10 3 cells/cm 2 and maintained in DMEM with 10% FBS at 37°C and 5% CO 2 for 4 hours.
  • adherent cells were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently stained with 5 ⁇ M l,l'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil) (Molecular Probes, Eugene, OR) in DMSO for 45 minutes at 37°C.
  • Dil l,l'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate
  • DMSO DMSO
  • Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO 2 . Cells were seeded onto the substrates at a density of 2.9 x 10 3 cells/cm 2 and maintained in DMEM with 10% FBS at 37°C and 5% CO 2 for 4 hours.
  • adherent cells were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently stained with 5 ⁇ M l,r-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil) (Molecular Probes, Eugene, OR) in DMSO for 45 minutes at 37°C.
  • Dil 5 ⁇ M l,r-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate
  • Example 29b (depending on substrate size) from random locations on each substrate using a Leica epifluorescent microscope equipped with a SPOT RT digital camera (Diagnostic Instruments, Sterling Heights, MI). The resulting images were quantified in terms of total projected cellular area using thresholding in MetaMorph. (Universal Imaging CorporationTM, a subsidiary of Molecular Devices Corporation, Downington, PA). The mean and standard deviation of the measurements are reported.
  • Example 29b Example 29b
  • Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO 2 . Cells were seeded onto the substrates at a density of 2.9 x 10 3 cells/cm 2 and maintained in DMEM with 10% FBS at 37°C and 5% CO 2 for 4 hours.
  • Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO 2 . Cells were seeded onto the substrates at a density of 2.9 x 10 3 cells/cm 2 and maintained in DMEM with 10%) FBS at 37°C and 5% CO 2 for 4 hours.
  • adherent cells were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently stained with 5 ⁇ M l,l '-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil) (Molecular Probes, Eugene, OR) in DMSO for 45 minutes at 37°C.
  • Dil 5 ⁇ M l,l '-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate
  • Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO 2 . Cells were seeded onto the substrates at a density of 2.9 x 10 3 cells/cm 2 and maintained in DMEM with 10% FBS at 37°C and 5% CO 2 for 4 hours.
  • adherent cells were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently stained with 5 ⁇ M l,r-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil) (Molecular Probes, Eugene, OR) in DMSO for 45 minutes at 37°C.
  • Dil 5 ⁇ M l,r-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate
  • Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO 2 . Cells were seeded onto the substrates at a density of 2.9 x 10 3 cells/cm 2 and maintained in DMEM with 10% FBS at 37°C and 5% CO 2 for 4 hours.
  • adherent cells were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently stained with 5 ⁇ M l,l '-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil) (Molecular Probes, Eugene, OR) in DMSO for 45 minutes at 37°C.
  • Dil 5 ⁇ M l,l '-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate
  • Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO 2 . Cells were seeded onto the substrates at a density of 2.9 x 10 3 cells/cm 2 and maintained in DMEM with 10% FBS at 37°C and 5% CO 2 for 4 hours.
  • adherent cells were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently stained with 5 ⁇ M l,l'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil) (Molecular Probes, Eugene, OR) in DMSO for 45 minutes at 37°C.
  • Dil l,l'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate
  • DMSO DMSO
  • Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO 2 . Cells were seeded onto the substrates at a density of 2.9 x 10 3 cells/cm 2 and maintained in DMEM with 10% FBS at 37°C and 5% CO 2 for 4 hours.
  • adherent cells were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently stained with 5 ⁇ M l,l '-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil) (Molecular Probes, Eugene, OR) in DMSO for 45 minutes at 37°C.
  • Dil 5 ⁇ M l,l '-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate
  • Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO 2 . Cells were seeded onto the substrates at a density of 2.9 x 10 3 cells/cm 2 and maintained in DMEM with 10%) FBS at 37°C and 5% CO 2 for 4 hours.
  • adherent cells were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently stained with 5 ⁇ M l,l'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil) (Molecular Probes, Eugene, OR) in DMSO for 45 minutes at 37°C.
  • Dil l,l'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate
  • DMSO DMSO
  • Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO 2 . Cells were seeded onto the substrates at a density of 2.9 x 10 3 cells/cm 2 and maintained in DMEM with 10%) FBS at 37°C and 5% CO 2 for 4 hours.
  • adherent cells were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently stained with 5 ⁇ M l, -dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil) (Molecular Probes, Eugene, OR) in DMSO for 45 minutes at 37°C.
  • Dil -dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate
  • DMSO DMSO
  • Each surface was modified for 24 hours in a solution of 1.0 mg/mL mPEG-DOPA 3 (or mPEG-OH as a control) at 50°C at the pH values shown in Figure 24.
  • a four-hour cell adhesion and spreading assay was conducted as described above in Example 9. Results are shown in Figure 24.
  • Cell adhesion resistance was conferred to all substrates treated with mPEG-DOPA 3 .
  • Cell adhesion and spreading on substrates treated with mPEG-OH did not differ from the unmodified surfaces (data not shown).
  • Example 31 Surfaces and Surface Preparation.
  • Silicon wafers (WaferNet GmbH, Germany) were coated with TiO 2 (20 nm) by physical vapor deposition using reactive magnetron sputtering (PSI, Villigen, Switzerland). Metal oxide coated wafers were subsequently diced into 1 cm x 1 cm pieces for ex-situ ellipsometry measurements.
  • Optical waveguide chips for OWLS measurements were purchased from Microvacuum Ltd. (Budapest, Hungary) and consisted of a AF45 glass substrate (8 x 12 x 0.5 mm) and a 200 nm-thick Sio .25 Tio .75 O 2 waveguiding surface layer.
  • TiO 2 -coated silicon wafers and waveguide chips were sonicated in 2-propanol for 10 minutes, rinsed with ultrapure water, and dried under a stream of nitrogen, followed by a 3 minute exposure to O 2 plasma (Harrick Scientific, Ossining, USA) to remove all organic components from the surface.
  • O 2 plasma Hard Scientific, Ossining, USA
  • waveguides were regenerated for reuse by sonication (10 minute) in cleaning solution (300 mM HCl, 1% detergent; Roche Diagnostics, Switzerland) and subsequent rinsing with ultrapure water to remove adsorbates.
  • XPS X-ray Photoelectron Spectroscopy
  • Measured intensities were converted to normalized intensities by atomic sensitivity factors, from which atomic compositions of surfaces were calculated. Average values obtained from three substrate replicates is reported in Tables 7-8. Standard deviations were typically ⁇ 10% of the mean and are omitted for clarity.
  • OWLS Optical Waveguide Lightmode Spectroscopy
  • the crude product was loaded onto silica gel and eluted with DCM, 5% methanol in DCM, 10% methanol in DCM, and 15% methanol in DCM.
  • the solvent was removed under vacuum to yield 5 as a white solid. The yield was 63%.
  • Precursor solutions of PEG-DA, 1, 7, and photoinitiator were prepared and mixed immediately before photopolymerization.
  • Stock solutions of PEG-DA (200 mg/mL) and! (40 mg/mL) were dissolved in N 2 -purged phosphate buffered saline (PBS, pH 7.4), where 7 (60 mg/mL) was dissolved in 50:50 PBS/95% ethanol previously purged with N 2 .
  • PBS N 2 -purged phosphate buffered saline
  • 7 60 mg/mL was dissolved in 50:50 PBS/95% ethanol previously purged with N 2 .
  • solutions of 1 or 7 were combined with PEG-DA to achieve a final concentration of PEG-DA and DHPD derivatives of 150 mg/mL.
  • the final VP concentration was adjusted to be between 135 and 300 mM.
  • the amount of DOPA incorporated into the photopolymerized gel was determined using a modification of the colorimetric DOPA assay developed by Waite and Benedict. Photocross-linked gels were stirred in 3 mL of 0.5 N HCI to extract DOPA monomers that were not incorporated into the gel network. 0.9 mL of the nitrite reagent (1.45 M sodium nitrite and 0.41 M sodium molybdate dihydrate) and 1.2 mL of IM NaOH were added to 0.9 mL of the extraction solution, and the absorbance (500 nm) of the mixture were recorded using a Hitachi U-2010 UV-Vis spectrophotometer with 2 to 4 minutes of NaOH addition. Standard curves were constructed using known 1 to 7 concentrations.
  • the other end of the cylinder was attached to a piezoelectric stepping motor (IW-701-00, Burleigh Instruments, NY) aligned in series with a 50 g load transducer (FTD-G-50, Schaevitz Sensors, VA) with a resolution of approximately 0.1 mN.
  • a fiber optic displacement sensor (RC100-GM2OV, Philtec, Inc., MD) measured the axial movement of the steel rod.
  • a TiO 2 -coated Si wafer was positioned below the hydrogel, and the Tio 2 surface was flooded with PBS in order to maintain the hydration of the gel. The indenter was advanced at 5 ⁇ m/s until a maximum compressive load of 4 mN was measured.
  • Elastic moduli were calculated by assuming Hertzian mechanics for the specific case of non-adhesive contact between an incompressible elastic hemisphere and a rigid plane, in which case the Hertzian relationship between load (E h ) and displacement ( ⁇ n ) becomes: 16R //2 E (1) 9 where R and E are the radius of curvature and the elastic modulus of the hemispherical gel, respectively. The radius of curvature of the gels was determined from height and width measurements obtained from a photograph of the gel.
  • Example 43 Chemical oxidation of PEG-DOPA into a hydrogel
  • Triethylamine (Et 3 N), hydrogen peroxide (30 wt%, H 2 O 2 ), sodium molybdate dihydrate, and sodium nitrite were purchased from Aldrich Chemical Company (Milwaukee, WI). L-Dopa was purchased from Lancaster (Windham, NH). 1-Hydroxybenzotriazole (HOBt) was obtained from Novabiochem Corp. (La Jo 11a, CA) and O-(Benzotriazol-l- yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HBTU) was acquired from Advanced ChemTech (Louisville, KY).
  • the solution was successively washed with saturated sodium chloride solution, 5% NaHCO 3 , diluted HCl solution, and distilled water.
  • the crude product was concentrated under reduced pressure and purified by column chromatography on Sephadex® LH-20 with methanol as the mobile phase.
  • the product was further purified by precipitation in cold methanol three times, dried in vacuum at room temperature, and stored under nitrogen at -20°C.
  • N-Boc-L-DOPA dicyclohexylammonium salt (5.9 mmoles), HOBt (9.8 mmoles), and Et ⁇ (9.8 mmoles) were dissolved in 50 mL of a 50:50 mixture of DCM and DMF.
  • HBTU (5.9 mmoles) in 25 mL of DCM was then added, and the reaction was carried out under argon at room temperature for 30 minutes. Recovery and purification of the product was performed as described above for I.
  • PEG-DOPA aqueous solutions were treated with nitrite reagent (1.45 M sodium nitrite and 0.41 M sodium molybdate dihydrate) followed by the addition of excess NaOH solution.
  • nitrite reagent (1.45 M sodium nitrite and 0.41 M sodium molybdate dihydrate)
  • the absorbance (500 nm) of the mixture was recorded using a Hitachi U-2010 UV/vis spectrophotometer, within 2 to 4 minutes of NaOH addition.
  • a standard curve was constructed using solutions of known DOPA concentration.
  • PEG-DOPA hydrogels [00258] To form PEG-DOPA hydrogels, sodium periodate (NaIO 4 ), horseradish peroxidase and hydrogen peroxide (HRP/H 2 O 2 ), or mushroom tyrosinase and oxygen (MT/O 2 ) were added to solutions of PEG-DOPA (200 mg/mL) in phosphate buffered saline (PBS, pH 7.4). For gelation induced by MT, the PBS was sparged with air for 20 minutes prior to adding MT. Gelation time was qualitatively determined to be when the mixture ceased flowing, as measured by inversion of a vial containing the fluid.
  • NaIO 4 sodium periodate
  • HRP/H 2 O 2 horseradish peroxidase and hydrogen peroxide
  • MT/O 2 mushroom tyrosinase and oxygen
  • Oscillatory rheometry was used to monitor the process of gelation and to determine the mechanic properties of the hydrogels.
  • Cross-linking reagent was added to aqueous solution of PEG-DOPA and the well-mixed solution was loaded onto a Bohlin VOR rheometer. The analysis was performed at a frequency of 0.1 Hz, a strain of 1%, and a 30 mm diameter cone and plate fixture with a cone angle of 2.5°.
  • DOPA-modified PEG was dissolved in 10 mM PBS solution (bubbled with argon for HRP/H2O2 and NaIO 4 or air for MT experiments). After adding the oxidizing reagent, the time-dependent UV/vis spectra of the solution were monitored at wavelengths from 200 to 700 nm at a scan rate of 800 nm/min. All samples were initially blanked against PBS buffer and recorded at room temperature using a Hitachi U- 2010 UV/vis spectrophotometer.
  • Molecular Weight Analysis [00261] Molecular weights were determined by GPC-MALLS on a DAWN EOS
  • PEG polyethylene glycol
  • Fmoc- PEG-NHS provides an amine for Boc- DOPA conjugation after Fmoc cleavage.
  • Piperidine (20% v/v in NMP) was used to deprotect Fmoc for 5 min and subsequently cantilevers were transferred to BOP/HOBt/DOPA (a molar ratio of 1 :1:1, final 8mM in NMP) solution with 10 ⁇ L DIPEA. The same procedure was used for Tyrosine modification.
  • Loading rate dependent force measurement revealed the energy landscape of DOPA binding (17).
  • the binding energy barrier is calculated by the force of logarithmic intercept at zero loading rate from the force transition occurred by pulling rate change and xb from the slope.
  • Silicon nitrite AFM cantilevers (Bio-Levers, Olympus, Japan) were used because of their small string constants (-5 pN/nm and -28 pN/nm).
  • XPS X-ray photoelectron microscopy
  • Silicon nitride surfaces (0.7 x 0.7 cm2) prepared in the high temperature chamber (ask to Keun Ho) were cleaned and modified as the same procedures described in AFM tip modification.
  • the photoelectron signal from carbon Is orbital was the major indicator for surface modification considering all abundant species of Si, O, and N in Si3N4 surfaces.
  • the present invention can be used with various other synthetic techniques well known to those skilled in the art to functionally modify a particular polymeric component for a subsequent coupling and preparation of the corresponding DOPA conjugate.

Abstract

Adhesive polymeric compositions which can comprise dihydroxyphenyl moieties and derivatives thereof, and related methods of use.

Description

POLYMERIC COMPOSITIONS AND RELATED METHODS OF USE
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application is a continuation-in-part of U.S. Serial No. 10/199,960 filed July 19, 2002 which claimed priority to U.S. Serial Nos. 60/306,750 and 60/373,919 filed, respectively, on July 20, 2001 and April 19, 2002. This application also claims priority to U.S. Serial No. 60/548,314 filed February 27, 2004, U.S. Serial No. 60/549,259 filed March 2, 2004. Each application is incorporated herein by reference in its entirety.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
[0002] The United States Government has certain rights to this invention pursuant to Grant Nos. DE13030, DE12599 and DE14193 from the National Institutes of
Health to Northwestern University and Grant No. NCC- 1-02097 from NASA to
Northwestern University.
BACKGROUND OF THE INVENTION
[0003] Mussel adhesive proteins (MAPs) are remarkable underwater adhesive materials that form tenacious bonds between mussels and the surfaces upon which the mussels reside. During the process of attachment to the surfaces, MAPs are secreted as fluids that undergo a crosslinking or hardening reaction which leads to the formation of a solid plaque. One of the unique features of MAPs is the presence of L-3,4- dihydroxyphenylalanine (DOPA), an unusual amino acid which is believed to be at least partly responsible for adhesion to substrates through mechanisms that are not fully understood. Mussels adhere to a variety of surfaces, including metal, metal oxide, polymers, plastics, and wood.
[0004] Control of cell and protein adhesion on surfaces is critical to the performance of biosensors, medical diagnostic products, any instrumentation and assays used requiring handling serum and other human/animal fluids, tissue engineering, localized in vivo drug delivery, implanted medical devices, healing of surgical incisions, adhesion of tissues such as bone and cartilage for healing, and nanotechnology (nanoparticle-based therapies and diagnostic tools). In many industrial applications, control of cellular and protein adhesion to surfaces is also important. Such applications include prevention of mussel attachment to boats and ships, piers, and other structures used in oceans and fresh water, prevention of algal and bacterial growth on water lines used for industrial and drinking water, and sensors used to measure water quality and purity.
[0005] In the medical arena, the physical or chemical immobilization of poly(alkylene oxides) (PAO), such as polyethylene glycol (PEG), polyethylene oxide (PEO), and PEO-PPO-PEO block copolymers, such as those available under the PLURONICS brand name, and polymers such as PEG/tetraglyme, poly(methoxyethyl methacrylate) (PMEMA), and Poly(methacryloyl phosphatidylcholine) (polyMPC) (E.W. Merrill, Ann. NY Acad. Sci., 516, 196 (1987); Ostuni et al., Langmuir 2001, 17, 5605-20, which are incorporated herein by reference) on surfaces has been employed as strategy to limit the adsorption of proteins and cells on surfaces. The methods currently employed to modify surfaces with polymers must be tailored for each type of material, and therefore require different chemical strategies. For example, noble metal surfaces, such as platinum, silver, and gold, can be modified using thiol (-SH) containing molecules, whereas metal oxides are often modified using silane coupling chemistry. No surface modification strategy exists that can be universally applied to different classes of materials. Moreover, many of the current methods rely on expensive instrumentation, complex synthetic procedures, or both.
SUMMARY OF THE INVENTION
[0006] The present invention are compositions which function e.g., as an adhesive, in a substantially aqueous environment. The preferred compositions generally comprise an adhesive moiety and a polymer moiety, the polymer moiety having a desired surface active effect (or other desired characteristics).
[0007] In one aspect, the adhesive moiety of a composition of this invention comprises dihydroxyphenyl derivatives including, di (DHPD) wherein the second DHPD is
Figure imgf000005_0001
i.e., a methylene derivative of dihydroxy phenyl. In yet a further aspect, the polymer moiety comprises poly (alkyleneoxide). In a very preferred practice, the adhesive moiety comprises DHPD, e.g., DOPA (discussed herein), and the polymer moiety comprises PEO-PPO-PEO block polymers (also discussed above).
[0009] In a further preferred practice the adhesive moiety comprises DHPD including a pendent chain comprising ethylenic or vinylic unsaturation such as, for example, an alkyl acrylate.
DETAILED DESCRIPTION OF THE INVENTION
[0010] Yet more specifically this invention comprises dihydroxyphenyl (DHPD) adhesivie compound of formula (I) wherein
Figure imgf000005_0002
Ri and R2 may be the same or different and are independently selected from the group consisting of hydrogen, saturated and unsaturated, branched and unbranched, substituted and unsubstituted C1-4 hydrocarbon;
P is separately and independently selected from the group consisting of -NH2, - COOH, -OH, -SH,
Figure imgf000006_0001
wherein Ri and R2 are defined above. a single bond, halogen,
Figure imgf000006_0002
wherein Ai and A2 are separately and independently selected from the group consisting of H, a single bond; a protecting group, substantially poly(alkyleneoxide), D
Figure imgf000007_0001
O wherein n ranges between 1 and about 3
Figure imgf000007_0002
R4 R4 is H, C|_6 lower alkyl, or poly(alkylene oxide)-C-C=CH2,
O R3
R3 is defined as above, and D is indicated in Formula (i). 1] In one aspect the poly(alkylene oxide) has the structure
Figure imgf000007_0003
wherein R3 and R4 are separately and independently H, or CH3 and m has a value in the range between 1 and about 250, A4 is NH2, COOH, OH, -SH, -H or a protecting group. [0012] In a very preferred form, DHPD is
Figure imgf000008_0001
Ri, R2, and P being defined as above.
[0013] In a further preferred form DHPD is of the structure:
Figure imgf000008_0002
wherein A2 is -OH and Ai is substantially poly(alkylene oxide) of the structure
Figure imgf000008_0003
R3, i and m being defined as in claim 2. Generally speaking the poly(alklene oxide) is a block copolymer of ethylene oxide and propylene oxide.
[0014] A method of this invention involves adhering substrates to one another comprising the steps of providing DHPD of the structure:
Figure imgf000009_0001
wherein Ri and R2 are defined as above; applying the DHPD of the above structure to one or the other or both of the substrates to be adhered; contacting the substrates to be adhered with the DHPD of the above structure therebetween to adhere the substrates to each other, and optionally repositioning the substrates relative to each other by separating the substrates and recontacting them to each other with the DHPD of the above structure therebetween.
[0015] In a preferred method, Ri and R2 are hydrogen. [0016] Definition: Dihydroxyphenyl derivatives (DHPD) for purposes of this application shall mean dihydroxyphenyl derivatives of the following structure:
Figure imgf000010_0001
wherein P, Ri and R2 are defined below, and n ranges between 1 and about 5. In one practice, Ri and R2 are hydrogen and P is, itself, dihydroxy phenyl. A preferred DHPD in a practice of the present invention is 1-3,4, dihydroxy phenyl alanine (DOPA), (generically),
Figure imgf000010_0002
wherein Ai and A2 are defined above. [0017] "Subtantially poly(alkylene oxide)" as used herein shall mean predominantly or mostly alkyloxide or alkyl ether in composition. This definition contemplates the presence of heteroatoms e.g., N, O, S, P, etc. and of functional groups e.g., -COOH, - NH2,-SH, as well as ethylenic or vinylic unstaturation. It is to be understood any such non-alkyleneoxide structures will only be present in such relative abundance as not to materially reduce, for example, the overall surfactant, non-toxicity, or immune response characteristics, as appropriate, or of this polymer.
BRIEF DESCRIPTION OF THE DRAWINGS
[0018] Figure 1 shows 1H NMR spectra of PLURONIC® F127, its carbonate intermediate (SC-PAO7) and DME-PAO7 in CDC13.
[0019] Figure 2 provides differential scanning calorimetry thermograms of 30 wt
% DME-PAO7, DOPA-PAO7, and unmodified PLURONIC® F127 aqueous solutions. Arrows indicate the location of gelation endotherm.
[0020] Figure 3 plots shear storage modulus, G', of a 22 wt % DME-PAO7 aqueous solution as a function of temperature at 0.1 Hz and a strain of 0.45%. Shown in the inset is the rheological profile of a 22 wt % unmodified-PLURONIC® F127 aqueous solution as a function of temperature.
[0021] Figure 4 plots shear storage modulus, G', of a 50 wt % DME-PAO8 aqueous solution as a function of temperature at 0.1 Hz and a strain of 0.45%. Shown in the inset is the rheological profile of a 50 wt % unmodified PLURONIC® F68 aqueous solution as a function of temperature.
[0022] Figure 5 plots storage moduli of DME-PAO8 aqueous solutions at 45 wt
% and 50 wt %, respectively, as a function of temperature at 0.1 Hz and a strain of 0.45%.
[0023] Figures 6A and 6B show differential scanning calorimetry thermograms of (A) DOPA-PAO7 and (B) DME-PAO7 at different concentrations upon heating. Arrows indicate the location of gelation endotherm observed only at higher polymer concentrations.
[0024] Figures 7A-C show high-resolution C(ls) XPS peaks for (A) un-modified
Au, (B) m-PEG-OH, and (C) m-PEG-DOPA. A dramatic increase in the ether peak at 286.5eV in (C) indicated the presence of PEG.
[0025] Figures 8A-C provide TOF-SIMS positive spectrum showing peaks representing catechol binding of gold. Spectra were normalized to Au peak (m/z ~ 197).
[0026] Figure 9 provides TOF-SIMS spectra showing the positive secondary ion peak at mass m/z ~ 43 for unmodified Au substrate, Au exposed to mPEG-OH, mPEG- DOPA powder and Au exposed to mPEG-DOPA.
[0027] Figure 10 shows TOF-SIMS spectra showing the positive secondary ion peaks for Au substrate chemisorbed with mPEG-DOPA. Catecholic binding of gold is observed at m/z ~ 225 (AuOC), 254 (AuOCCO), and 309. Less intense AuOaCb peaks are seen at m/z ~ 434, 450, 462, and 478. The periodic triplets seen in the m/z range 530- 1150 correspond to Au bound to DOPA-(CH CH2O)n, where each subpeak is separated by 14 or 16 amu, representing CH2, CH2CH2, and CH CH2O in the PEG chain. This pattern was observed for n = 1-15.
[0028] Figure 11 shows SPR spectra of protein (0.1 mg/ml BSA) adsorption onto modified and unmodified gold surfaces. mPEG-DOPA and mPEG-MAPd modified surfaces exhibited reduced protein adsorption compared to bare gold and mPEG-OH modified surfaces.
[0029] Figure 12 shows mPEG-DOPA concentration dependence of anti-fouling behavior. Gold surfaces were modified for 24h at the mPEG-DOPA concentrations indicated, followed by analysis of the density and area of attached cells. (* = p<0.05, ** = pO.Ol, *** = pO.OOl; black bars^total proj. area, gray bars=surface cell density)
[0030] Figure 13 compares cell attachment and spreading on bare gold, mPEG-OH-treated gold, and gold modified with mPEG-DOPA 5K, mPEG-MAPd 2K, and mPEG-MAPd 5K under optimal conditions (50mg/ml for 24h). (black bars=total proj. area, gray bars=surface cell density; *** = pO.OOl) [0031] Figures 14 A-C are a series of SEM micrographs indicating the morphology of NIH 3T3 fibroblasts on (A) unmodified Au, (B) Au treated with mPEG-OH, and (C) mPEG-DOPA-modified Au. All treatments were at 50mg/ml in DCM for 24 h.
[0032] Figure 15 shows the UV/vis absorption spectrum of mPEG-DOPA stabilized magnetite nanoparticles suspended in several aqueous NaCl solutions at the concentrations as shown and plotted therein. Addition of NaCl did not induce nanoparticle precipitation.
[0033] Figure 16 shows addition of salt to untreated Au nanoparticles induces aggregation. Shown are UV/vis scans of lOnm untreated Au nanoparticles suspended in aqueous NaCl solutions (concentrations as shown and plotted therein). The attenuation and shift of the 520 nm absorption band with increasing NaCl concentration reflects aggregation of the nanoparticles.
[0034] Figure 17 illustrates addition of salt to mPEG-DOPA stabilized Au nanoparticles does not induce aggregation. Shown are UV/vis scans of lOnm mPEG- DOPA stabilized Au nanoparticles suspended in aqueous NaCl solutions (concentrations as shown and plotted therein). The lack of attenuation and shift of the 520nm absorption band with increasing NaCl concentration reflects effective stabilization of the nanoparticles.
[0035] Figure 18 plots the UV/vis absorption spectrum of mPEG-DOPA stabilized CdS nanoparticles suspended in aqueous NaCl solutions (concentrations as shown and plotted therein.
[0036] Figure 19 (plots XPS survey scans of unmodified TiO2 and TiO2 treated with mPEG-DOPAι-3.
[0037] Figure 20 plots the long-term resistance to cell adhesion on TiO2 and TiO2 modified with mPEG-DOPAι-3. The duration of the non-fouling response is proportional to the length of the DOPA peptide anchoring group. Adherent cells were visualized with calcium AM. [0038] Figure 21 plots the high-resolution XPS scans of the Cis region of TiO2 substrates modified with mPEG-DOPA1-3. Of note is the increase in the ether carbon peak (286. OeV) with increasing length of the DOPA peptide anchor.
[0039] Figure 22 plots the high-resolution XPS scans of the Ols region of TiO2 substrates modified with mPEG-DOPAι-3. The peak at 532.9eV representing polymeric oxygen increases while the Ti-O-H peak (531.7eV) decreases with increasing DOPA peptide length.
[0040] Figure 23 plots the results of the Robust Design experiment on 316L stainless steel,
[0041] Figure 24 plots the 4-hour cell attachment to a variety of surfaces modified by mPEG-DOPAι-3 using a 24-hour modification at 50°C at the indicated pHs.
[0042] Figure 25 plots the % gel conversion versus the UV exposure time in minutes.
[0043] Figure 26 plots the mole fraction of DOPA incorporated versus the mol % of 1 or 7 in the precursor solution.
[0044] Figure 27 plots the % gel conversion versus mol % 1 or in the precursor solution.
[0045] Figure 28 X-ray Photoelectron Spectroscopy XPS analysis of a silicon nitride surface.
[0046] Figure 29 is a free monitoring of functionalized silicon nitride cantilevers.
[0047] Figure 30 is an analysis of entropic elasticity of poly(ethylene glycol).
[0048] Figure 31 is a force measure of side chain modified DOPA.
[0049] Figure 32 is a proposed model of DOPA-T1O2 binding mechanism.
[0050] Figure 33 is an atomic force microscopy arrangement.
[0051] Figure 34 is data regarding force measurement. [0052] Figure 35 is adhesion data.
[0053] Figure 36 is synthetic route and data analysis.
[0054] These dihydroxyphenyl derivative ("DHPD") adhesives function in an aqueous environment. To form the polymeric composition, a DHPD moiety which generally provides adhesive functionality coupled to a polymer which provides the desired surface active effect. These components will be described in more detail below.
[0055] These adhesives and polymeric compositions have many uses, including prevention of protein and/or cell adhesion to a surface in various medical, industrial and consumer applications. The DHPD adhesives can also be used as substitutes for sutures for a wound and as aids in healing bone fractures or cartilage-to-bone damage. These and other uses will be described in more detail below.
[0056] The preferred polymer compositions of the present invention have the following structure:
Figure imgf000015_0001
wherein, for each compound of formula (la), Ri and R2 are defined separately and independently as above,
Pi and P2 are separately and independently defined as P in formula (I);. n and m independently range from 0 to about 5, provided that at least one of n or m is at least 1;
[0057] Adhesive Moiety The adhesive moiety of the present invention is a dihydroxyphenyl derivative ("DHPD") having the following preferred structure:
Figure imgf000016_0001
wherein Ri, R2 and P are defined above and t ranges between 1 and about 10, preferably about 1 to about 5 and most preferably 1 to about 3. The DHPD adhesive can function in an aqueous environment. In this context, an aqueous environment is any medium comprising water. This includes without limitation water, including salt water and fresh water, cell and bacterial growth media solutions, aqueous buffers, other water-based solutions, and body fluids. The DHPD moiety can be derivatized. As would be understood by those skilled in the art, such derivatization is limited by the retention of the desired adhesive characteristic.
[0058] Polymeric Component
Various polymeric components providing a surface active effect and other desired characteristics will be well-known to those skilled in the art made aware of this invention. The desired surface active effect relates to reduced particulate agglomeration and anti-biofouling, including resistance to cell and/or protein adhesion. For instance, the polymer component can be water soluble, depending upon end-use application, and/or capable of micelle formation depending upon various other end-use applications. Polymers useful in the present invention include, but are not limited to, polyethylene glycol (PEG), polyethylene oxide (PEO), polypropylene oxide (PPO), PEO-PPO-PEO block copolymers, polyphenylene oxide, PEG/tetraglyme, PMEMA, polyMPC, and perfluorunated polyethers.
[0059] The polymeric compositions can be synthesized in several ways. For example, the polymeric compositions may be synthesized through a general synthetic procedure for polymer end-group activation. Various polymers or monomeric components thereof can be activated using carbonate chemistry. In particular, a succinimidyl carbonate- activated polymeric component reacted with DHPD moiety can provide a stable urethane conjugate. Two of the many possible pathways (a) and (b) in Scheme la and lb, below, show coupling with a poly(alkylene oxide) in either aqueous or non-aqueous solvents, without compromising desired bioadhesion. For instance, a DHPD residue can be coupled to a polymeric component to provide the desired conjugate composition, through either urethane or amide bond formation. These synthetic counts are shown in Scheme la and lb which are discussed in greater detail below.
CH3 HO(CH2CH20)m— (CHCH20)n-(CH2CH20)m-OH
Figure imgf000018_0001
Et3N, DMAP, anhydrous conditions
Figure imgf000018_0002
Figure imgf000018_0003
DME-PA07 m=100, n=65 DME-PA08 m=78, n=30
SCHEME la
Figure imgf000019_0001
2) Acidified by HCI to pH=2.0
Figure imgf000019_0002
HOO
Figure imgf000019_0003
DOPA-PA07 m=100, n=65 D0PA-PA08 m=78, n=30
SCHEME lb [0060] More particularly, if coupled to the polymeric component via urethane bond formation, a carboxylic acid group of the DHPD component can be esterified or derivatized with various other functional groups. Alternatively, the DHPD component can be coupled to a polymeric component (e.g., amidation or esterification depending on polymer end group, -NH2 or -OH) providing a DHPD functionality which can be derivatized by any of numerous known protecting groups, including without limitation Boc, Fmoc, borate, phosphate, and tributyldimethylsilyl. N-group protection of a DHPD component can leave the carboxylic acid group available for multi-functional derivatization and/or a higher density of polymeric components conjugated therewith. [0061] Accordingly, in part, the present invention is also a method of using urethane synthesis to incorporate a DHPD residue into a polymeric system. Such a method includes (1) providing a polymeric component terminating in a plurality of monomers, each having a functional end group; (2) preparing a carbonate derivative of the polymeric component; and (3) preparing a urethane moiety upon reaction of the carbonate derivative and at least one DHPD moiety. As described above, a polymeric component utilized in conjunction with this method can include those having terminal monomeric functionality reactive with a reagent providing the desired carbonate derivative and, ultimately, providing a urethane moiety coupling the polymeric and DHPD components. Various other coupling reagents and/or hydroxy-terminating polymeric components can be used to provide the desired urethane moiety.
[0062] In part, the present invention is also a method of using a carbonate intermediate to maintain catecholic functionality of a DHPD-incorporated polymeric composition and/or system, or to otherwise enhance the adhesion properties thereof. Such a method includes (1) providing a polymeric component terminating in a plurality of monomers each having a functional end group; (2) reacting the polymeric component with a reagent to provide a carbonate intermediate; and (3) reacting the carbonate intermediate with at least one DHPD moiety. Without limitation to any single theory or mode of operation, this inventive method can be considered a way of enhancing the reactivity of the polymeric component end group, via a suitable carbonate intermediate. Subsequent reaction at the amino-nitrogen of DHPD moiety provides the corresponding conjugate while maintaining catecholic functionality.
[0063] In accordance with this invention, as demonstrated in Scheme la, various synthetic routes can be used to couple DHPD moieties to such carbonate activated intermediates. DOPA methyl ester (DME), prepared by the reaction of DOPA with methanol in the presence of thionyl chloride, can be used in organic solvents. Reaction progress can be monitored by TLC and NMR, with the coupling reaction virtually complete in one hour (with representative conjugates DME-PAO7 (from PAO PLURONIC® F127) and DME-PAO8 (from PAO PLURONIC® F68)). High product yields were obtained upon purification from cold methanol.
[0064] The free carboxylic form of DOPA can be coupled with the carbonate intermediate in alkaline aqueous solution. It is well known that the chief difficulty in working with DOPA is its ease of oxidation (to DOPA-quinone and other products), which readily occurs in alkaline aqueous solutions. To prevent unwanted oxidation of DOPA catechol side chains during coupling under alkaline conditions, a borate-protected DOPA can be first formed by adding DOPA to aqueous sodium borate (Scheme lb). The resulting complex is remarkably stable in neutral or alkaline solutions, and can be readily deprotected under acidic conditions. Taking advantage of complexation between DOPA and borate, DOPA was coupled to the ends of several commercially-available PAOs under alkaline aqueous conditions to yield DOPA-PAO7 and DOPA-PAO8. Visual inspection of the reaction solution revealed the absence of strongly absorbing DOPA-quinone, an indication that DOPA remains unoxidized during the reaction. At the completion of the reaction, acidification with HCI resulted in deprotection of the DOPA endgroups of the block copolymer.
[0065] Both 1H NMR spectra and colorimetric assay confirmed the compositions of the succinimidyl activated reaction intermediates and all four DOPA-modified PAOs of Scheme 1. Shown in Figure 1 are 1HNMR spectra of PAO PLURONIC® F127, the succinimidyl carbonate activated intermediate (SC-PAO7), and the corresponding DOPA methyl ester modified PAO (using PLURONIC® F127, DME-PAO7). The sharp peaks at —2.8 ppm due to the -CH2- protons from the succinimidyl carbonate group and at ~4.4 ppm due to the -CH2-0- protons from the only ethylene oxide group adjacent to the carbonate group in activated-PAO completely disappear from the XΗ NMR spectra of the DOP A-containing PAO, whereas a series of new peaks appear due to the introduction of DOPA moieties into the copolymers. One characteristic feature of the 1H NMR spectra of the DOP A-containing PAO is the appearance of one singlet and two doublets in the range of 6.5-6.9 ppm corresponding to the three protons on the DOPA phenyl ring. Similar features were also observed in the 1HNMR spectrum (not shown) of the DOPA- PAO conjugate synthesized from aqueous solution.
[0066] Based on the assumption of two available succinimidyl carbonate groups in the corresponding carbonate intermediates, SC-PAO7 and SC-PAO8, coupling efficiencies of DOPA methyl ester and DOPA to these two PAOs were quantitatively found to be in the range from 76% to 81% as obtained from colorimetric analysis (Table 1). The reported coupling efficiencies are the average values of at least three repeated syntheses performed under the same conditions and were not found to increase significantly when a larger excess of DOPA was used in the reaction. Similar coupling efficiencies were also found for DOPA-PAO7 and DOPA-PAO8 made from aqueous solutions, suggesting that the hydrolysis of succinimidyl carbonate activated PAOs is slow in the aqueous alkaline solution containing Na2B4O7.
[0067] In contrast to coupling efficiencies, the product yields (shown in Table 1) of selected DOPA-modified PAOs synthesized in aqueous solution were found to be lower than those synthesized in organic solvent. This may be due to the surfactant properties of the starting PAO material, causing the low efficiency of extraction of DOPA-modified PAO with dichloromethane from water. It should be noted that the free carboxylic acid in DOPA-PAO7 and DOPA-PAO8 can be further functionalized using standard peptide chemistry to tailor the properties of the block copolymers. The four DOPA-modified PAOs of Table 1 could be stored at -20°C indefinitely with no discoloration or change in properties. Table 1 Coupling efficiency and product yield of DOPA modified PLURONIC® Coupling Efficiency (%)* Product Yield (%) DME-PAO7 78.0 ± 4.0 75.0 ± 5.0 DOPA-PAO7 80.0 ± 4.0 52.0 ± 3.0 DME-PAO8 76.0 ± 2.0 76.0 ± 4.0 DOPA-PAO8 81.0 ± 2.0 49.0 ± 2.0
*Determined by colorimetric analysis as taught by Waite and Benedict (Waite, J.H. & Benedict, C. V. Assay of dihydroxyphenylalanine (dopa) in invertebrate structural proteins. Methods in Enzymology 107, 397-413 (1984), which is incorporated herein by reference). [0068] Control of cell and protein adhesion on surfaces is critical to the performance of biosensors, medical diagnostic products, any instrumentation and assays used requiring handling serum and other human/animal fluids, tissue engineering, localized in vivo drug delivery, implanted medical devices, healing of surgical incisions, adhesion of tissues such as bone and cartilage for healing, and nanotechnology (nanoparticle-based therapies and diagnostic tools). In many industrial applications, control of cellular and protein adhesion to surfaces is also important. Such applications include without limitation prevention of mussel attachment to boats and ships, piers, and other structures used in oceans and fresh water, prevention of algal and bacterial growth on water lines used for industrial and drinking water, and sensors used to measure water quality and purity. [0069] The polymeric compositions of the present invention can be used as coatings to prevent protein and cellular adhesion to devices for medical and research applications. These include without limitation such uses as coatings for medical implants, coatings for surgical devices, coatings for devices that handle serum and other animal or human derived materials, medical diagnostic devices, and biosensors. Alternatively, the polymeric compositions can be tissue adhesive polymeric hydrogels for medical uses such as tissue sealants, gels for prevention of surgical adhesion (scar tissue formation), bone and cartilage adhesives, tissue engineering, and site specific drug elution and for research uses such as immobilization of proteins including antibodies and small molecule analytes including pharmaceuticals. In addition, there are various industrial and consumer product uses of these coatings and hydrogels including without limitation prevention of marine biofouling (attachment of algae, bacteria, and mussels to surfaces underwater), prevention of bacteria contamination of water streams to industrial plants such as electronic and drug manufacturers, prevention of bacterial contamination of drinking water streams, dental and denture adhesives, underwater adhesives to deliver indicators, coatings for water purity and measurement sensors, paints used for prevention of biofouling, and use in cosmetics to adhere desired fragrances and colorants to hair, eyelids, lips, and skin, to form temporarily skin coloring such as tattoos and the like, and for resealable adhesives for consumer products such as storage bags. The present methods can be used to prepare a variety of polymer modified surfaces for both medical (diagnostics, devices, nanoparticle-based therapies) and nonmedical (paints and other particle dispersions, MEMS, quantum dots, nonfouling surfaces) technologies. [0070] Adhesive hydrogels can be also formed using the present methods. The DHPD adhesive is attached to polymers capable of forming a hydrogels in vivo or in vitro. These hydrogels can be formed by a number of methods including the use of self- assembling polymers that form gels at higher temperatures such as normal human body temperatures, the use of polymers that can be cross-linked by an enzymatic reaction, the use of polymers that can be subjected to oxidation to form cross-linked hydrogels, and the use of polymers that can be subjected to photoactivation to produce cross-linked hydrogels. Anti-Biofouling Coatings
[0071] The anti-biofouling coatings of the present invention may be applied to medical devices, such as vascular or arterial stents, pacemakers, heart valves, glucose monitors and other biosensors, vascular wraps, defibrillators, orthopedics devices, and surgical devices, including sutures and catheters. The polymeric compositions of the present invention can be used as coatings to prevent protein and/or cellular adhesion to a device for medical and research applications. These include without limitation such uses as coatings for medical implants, coatings for surgical devices, coatings for devices that handle serum and other animal or human derived materials, medical diagnostic devices, and biosensor. Among the challenges in modifying biomaterial surfaces with polymers for cell adhesion resistance are producing a sufficiently high density of polymer capable of repelling proteins and cells and producing a coating that cover the surface completely. This is particularly a problem with devices containing multiple components made of different materials. A surface may be modified by the polymeric composition of the present invention in any number of ways. For example, the polymeric composition may be absorbed onto the surface or a DHPD moiety containing a polymerization initiator may be adsorbed onto the surface and polymer growth initiated from the surface. With the latter, a number of polymerization techniques are possible, including without limitation surface initiated radical polymerization, radical polymerization methods, ionic polymerization, ring-opening polymerization, and photopolymerization.
[0072] For example, by utilizing the unique solubility properties of PEG, the surface density of polymer can be increased by treating surfaces with PEG solutions near the lower critical solution temperature (LCST), or cloud point. While not wanting to be bound by theory, applicants believe that under the high ionic strength and elevated temperature conditions used in the present invention, PEG molecules have a reduced hydrodynamic radius, which in principle allows a higher density of PEG chains to pack on a surface than under standard conditions. This approach is useful for polymers that show inverse solubility transitions at high temperature and high ionic strength, such as poly(ethylene glycol), poly(N-isopropylacrylamide) and other N-substituted poly(acrylamides) that show inverse solubility transitions.
[0073] By modifying surfaces with various polymeric composition of the present invention, resistance to cell and protein attachment is conferred for up to 7 days, 14 days, 21, days, 30 days, 60 days, 90 days and 120 days or longer. The number of DHPD moieties in the adhesive component and the pH of the modification buffer are responsible for most of the variation in the cell and/or protein adhesion resistance of the modified materials. For those surfaces that are modified by adsorption, adsorption time and polymeric composition concentration contributes little to the variation in cell and/or protein adhesion resistance of the modified materials. The greater the number of DHPD moiety monomers in the adhesive component, the better the cell and/or protein adhesion resistance. The density of the polymeric composition on the surface correlates well with resistance to cell and/or protein adhesion. The thickness of the coating layer can be from about 20 A to about 100 μm, including 30 A, depending on the polymer composition used and the pH of the modification buffer.
[0074] The concentration of the polymer composition used for modification of a surface can be from about .1 mg/ml to about 75 mg/ml. The pH of the modification buffer can be from about 3 to about 9. The modification time can be from about 10 minutes to about 72 hours. The temperature of the modification can be from about 25°C to about 60°C.
[0075] As shown in Figure 19, XPS survey scans of unmodified TiO2 revealed strong peaks at -458 eV (Ti2p) and -530 eV (Ols) characteristic of native oxide, as well as a small peak at 248.7 eV (Cis) as a result of adventitious hydrocarbon contamination. TiO2 substrates treated with mPEG-DOPA1-3 under cloud point conditions, however, demonstrated dramatic increases in surface-bound carbon as reflected by the Cis peak, suggesting the presence of PEG on the surface. Moreover, the increases in the Cis peaks observed after modification with mPEG-DOPAι-3 were directly proportional to the number of terminal DOP As present. Additionally, a small peak at 400 eV (Νls) was seen in the spectrum of the TiO2 surface modified with mPEG-DOPAι-3, representing the amide nitrogen in DOPA.
[0076] Quantitative analysis of the high resolution XPS data of the substrate surfaces can provide useful information on relative amounts of PEG bound to the surface. Table 2 shows the titanium, oxygen, and carbon atomic composition calculations for TiO2- modified with mPEG-DOPAι-3. The oxygen signal is further subdivided into metal oxide (Ti-O-Ti), surface hydroxide (Ti-O-H), and organic oxygen and coupled water (C- O, H2O) species. Table 2 Titanium, oxygen, and carbon atomic composition calculations
Figure imgf000026_0001
aTrace amounts of N were neglected. bAssumed to be water bound to the surface.
[0077] The ratio of Ti to Ti-O-Ti for all substrates differs substantially from the theoretical stoichiometry of 2.0; the difference is likely due to a sampling depth that goes beyond the depth of the surface oxide (3-4 mm). This result is expected given the Flory radius of a 5000 Mw PEG (2.8 mm) and a typical XPS sampling depth of 5-10 mm. On surfaces modified with mPEG-DOPA1-3, the Ti-to-C atomic ratio decreased dramatically with increasing DOPA peptide length, corresponding to increases in the amount of adsorbed PEG. The observed C to organic oxygen (C-O) ratio exceeds the theoretical value of 2.0 for pure PEG, which suggests that adventitious hydrocarbon contamination remains on modified surfaces. These results are shown in Table 3. Table 3 Atomic ratio for adsorbed polymeric compositions
Figure imgf000026_0002
[0078] DOPA creates strong, reversible bonds with TiO2. The energy of the bond is 30.56 kcal/mol and needs about 800 pN to be detached from TiO2 at the single molecule level, which is four times stronger than the interaction between Avidin and Biotin. The DOPA-TiO2 strength of interaction is about midway between that of Avidin-Biotin, one of the strongest hydrogen bond based interactions in biology (0.1-0.2 nN) and a covalent bond (>2nN).
[0079] To study DOPA adhesion properly, the following conditions were used: single molecule approach, an aqueous environment, and a platform for DOPA immobilization. Atomic Force Microscopy (AFM) was chosen as a tool for the investigation which satisfies those three conditions and is sensitive enough to measure the viscoelastic properties of soft materials: protein, DNA, and synthetic polymers at a single molecule level. Amine moieties were introduced to a cantilever tip (Si3N4) and then the .mixture of methoxy-poly(ethylene glycol, mPEG) and Fmoc-terminated PEG (Fmoc-PEG) derivatives were conjugated. (Boc-)DOPA was coupled to the amine groups generated by the cleavage of Fmoc (Figure 33C). A 5-10 molar excess of mPEG to DOPA-PEG was used so that single, immobilized DOPA4 PEG could be isolated. This molecular configuration sterically hindered the molecular dynamics of DOPA-PEG, thus providing an explanation for the result of DOP Aoxidation experiments later (Figure 36). Chemical reaction steps were monitored by X-ray photoelectron spectroscopy (XPS) showing successful PEG modifications on flat silicon nitride surfaces (lxlcm2) (Fig 28). Chemical groups introduced onto silicon nitride tip surfaces change electrostatic properties, which are also good indicators of surface modifications (Fig 29- A). It is important to note that the difference of approaching signals was detected between bare and modified cantilevers representing resistance force due to molecular layers (Fig 29- B).
[0080] DOPA conjugated cantilevers displayed significant adhesion accompanied by entropic elasticity of the PEG chain (Fig 34). A histogram of the force distribution shows a uni-modal shape indicating only single adhesive event, which is different compared to the case of a multivalent protein, Avidinl2. Force-Distance (F-D) measurements were collected and a statistical analysis performed (Fig 34, n= 105). The average force was 785 pN in water at a loading rate of 180 nN/s. Most importantly, the length of stretched PEG (36 nm) was consistent with the expected contour length of a PEG molecule (37 nm, Fig 30). These data are observing a single molecular event: monovalent binding of DOPA, polydispersity of PEGs, and tip geometry. A single DOPA was conjugated at the end of a polymer chain, to serve as one binding unit on a TiO2 surface (Fig 33). This monovalency is different from metal- Histidine6 (Metal- (His)6) studies where tethered (His)6 provides three metal chelating sites (3 x metal/(His)2)13-15. In addition, a 'mountain-tree' like configuration of PEG5 antilever can separate two different DOPA detachment signals because of polydispersity of PEGs (tree) and the spherically-shaped tip (mountain) (r = 25 nm).
[0081] Defined, "d" is the z-displaced distance of piezoelectric device when a single DOPA-PEG molecule was fully stretched during retraction (Fig 34C). The 'd' values appeared to be almost constant throughout many repeated cycles although it did vary slightly (Fig 34A). This small variation might be due to DOPA binding to the surface at different angles. An important feature of our experiment is that the unbinding signals are epetitions using the 'identical' DOPA molecule. This is compared to the traditional approach of single molecule pulling experiments where tips picked up one molecule randomly. This also demonstrates that the DOPA adhesion chemistry was completely reversible. This reversibility led us to the conclusion that the weakest chemical linkage from substrate to tip (TiO2~Si3N4) is the Ti(surface)-O(DOPA) bond. [0082] The result suggested that at -0.8 nN, the DOPA-ΗO2 interaction is mechanically midway between that of Avidin-Biotin, one of the strongest hydrogen bond-based interaction in biology (0.1-0.2 nN) and covalent bond (>2 nN). Energy information from force data by changing the loading rate, the amount of force applied per unit time. Changes over four orders of magnitude of a loading rate generated four different force distributions to map the energy landscape of DOPA binding. Linear-log line plot of force vs. loading rate in Fig 34D provided binding energy and a distance after which the association is removed along the applied force direction. DOPA had an energy barrier of 28.1 kcal/mol, and the distance needed to reach the activation energy maximum was 1.27 A (Fig 34E).
[0083] The binding orientation of DOPA is believed to be with the two hydroxyl groups on the aromatic ring pointed down toward the surface. Therefore, the confirmation of chemical groups responsible for single molecule adhesion signals in AFM is important to exclude other binding chemistry due to different orientations. Two methods were used. [0084] First, chemical modification of the hydroxyl group covalently by ertarybutyldimethylsiloxane, (TBDMS) resulted in no binding during two hundred approach-retraction cycles (Fig 31 first line). However, deprotection of the TBDMS groups regenerated the binding ability of DOPA (Fig 31, bottom two lines). [0085] Second, the protection by ionic complexation with borate completely suppressed the strong adliesion of DOPA as well (Fig 31, n=200). These data clearly confirmed the di-hydroxyl group of DOPA as the true structural source for strong and reversible binding.
[0086] Mussels developed an interesting way to create such a strong binding in water, a post-translational modification of tyrosine by tyrosine hydroxylase. This enzyme catalyzes a reaction of adding one hydroxyl group using tyrosine as a substrate and a large amount is found in threads and plaques where DOPA exists as well. It is surprising to say that the small post-translational modification (-OH) seems to produce a huge change of adhesive ability. Thus, experiments were designed to show a correlation between the posttranslational modification and binding ability.
[0087] A tyrosine tethered cantilever was prepared instead of DOPA, and tyrosine adhesion on TiO2 was investigated. No detectable force signals were observed except some non-specific adhesions with low probability (Fig 35A). To reject the hypothesis that the tip used in this experiment didn't have any tyrosine molecules, the TiO2 surface was replaced with gold. The aromatic ring of tyrosine binds to a gold surface in a parallel orientation to the surface through π-π electron interaction, which is a well-known mechanism in surface adsorption chemistryl8,19. The same cantilever used in TiO2 produced relatively strong adhesions repeatedly on gold surfaces (Fig 35B). The statistical analysis of force distribution showed the π-π electron binding strength of 398 (± 98) pN which was about 50% strength compared to the DOPA-TiO2 interaction (Fig 35C). The force signals from tyrosine-gold binding also exhibited the same characteristics as previously shown in DOPA-ΗO2 interaction: elastic stretching of PEG with an expected contour length and repetitive signal appearance with similar 'd' (Fig 33C). In this experiment, it was clearly proved that the tyrosine hydroxylase-mediated post-translational modification greatly improved the binding ability of DOPA from almost zero to 800 pN.
[0088] Biological roles of DOPA go beyond adhesiveness upon oxidation: it crosslinks polypeptide chains resulting in stiffer materials found throughout threads and pads. The crosslinking mechanism has multiple pathways starting from a chemically unstable DOPAquinone structure. Aryl-aryl ring coupling (di-DOPA) has been found in mussel adhesive proteins20 but Michael addition (quinone-alkylamine adducts) products have been found in other species not mussels (Fig 36A). Therefore, these structures may occur as results of oxidation in mussels as well. It is clear in terms of crosslinking but is under debate with respect to adhesive properties after maturation i.e. oxidation. It was demonstrated that the DOPAquinone structure is not a major player for adhesiveness. The DOPAquinone-PEG chain is spatially and chemically stabilized by excess co- conjugation of methoxy-PEG molecules (5-10 molar equivalent) which is an important molecular configuration for preventing further reactions of DOPAquinone. [0089] Time-resolved monitoring force signals of single DOPAquinone triggered by increasing pH (=9.7) uncovered interesting things that have been hidden so far. First, measured AFM signals showed two clear distributions in terms of force magnitude: high force and low force (Fig 36B). Statistical analysis of the data yielded two clear histograms with 178 ± 62 pN for low and 741 ± 110 pN for high force (Fig 36C). The quinone binding can be assigned to the low force region because it appeared only after the oxidation was triggered and subsequently became more frequent over time (Fig 36D). The slow kinetic feature of DOPA oxidation contributed to initial high frequency of DOPA signals. This is the first single molecule experiment about detecting the structural change of a small molecule upon external stimulus. Based on these results, the possibility of the DOPAquinone structure being responsible for the high adhesiveness can be ruled out. Therefore, without being bound to any theory, the regeneration of reduced form i.e. di-hydroxyl group of DOPA during oxidation, is believed to be a very important requirement for maintaining or changing adhesive properties of DOPA containing materials at an interface.
[0090] In addition, the DOPA anchoring system can be a new platform to study other extensible biological macromolecules such as polysaccharides, DNA, and proteins. In the study performed, it already presented the elastic property of PEG (Mw 3400) and is believed to be the shortest chain length ever studied until now. This could be achievable simply because two defined anchoring methods were used at both ends: (1) covalent bonds between PEG and cantilever and (2) DOPA anchoring between PEG and substrate. This method is also highly contrasted with the conventional single molecule experiments where a tip 'sees' different molecules at every single movement of a cantilever. It has been a big barrier to investigate molecular responses upon external stimuli if a given stimulus was not hundred percent effective23,. The DOPA-based anchorage system can be an alternative technique to overcome these problems in current single molecule pulling experiments.
[0091] Currently, there is no clear answer why DOPA behaves like a reversible glue similar to 'Post-It'. Two molecular binding models, nomonuclear bidentate (Fig 32A, right) and binuclear bidentate (Fig 32A, left), are available but both did not consider the reversible binding of DOPA because the studies focused on only adsorption processes not desorptioms. Therefore, the nature of chemical binding was assumed to be mainly covalent independent of factors resulting from the removal of water molecules after adsorption. One study using FTIR suggested that the nature of DOPA-ΗO2 binding may be 60% ionic and 40% covalent. Based on this finding, a the molecular adsorption models were revised to incorporate the reversibility where multiple hydrogen bonds are formed in water (Fig 32B)
Figure 33 An experimental design and a single molecular DOPA adhesion
[0092] A picture describes how the blue mussel (Mytϊlus Edulis) sticks to metal oxide surfaces. The circle included one plaque where the unusual amino acid, DOPA, was found.
[0093] (B) Two major protein components uncovered in plaques in mussels, Mefp-3 and Mefp-5. These mussel adhesive proteins have high content of DOPA: 27 mole % of Mefp-5 and 21 % of Mefp-3. Bold Y (Y): DOPA, Italic S (S): phospho-serine, Italic R(R): hydroxy arginine.
[0094] (C) AFM tip modifications. Polymerization of 3- aminopropyltrimethoxysilane (APTMS) introduced amine groups on Si3N4 tip surfaces (not drawn). A long chain describes a PEG molecule conjugated with single (Boc)-DOPA at the end. Mixture of mPEG-NHS(2k) and Fmoc-PEG-NHS(3.4k) at a molar ratio of 5-10:1 was used to stabilize DOPA-PEG molecule (see supplemental section for details).
Figure 34. Single molecule force measurement and determination of energy landscape of DOPA binding on TiO2 surfaces
[0095] Four representative AFM single molecule DOPA detachment signals from one cantilever. Those four signals were not consecutively generated (signals were deleted which didn't show any adhesions). Despite a low probability to detect DOPA adhesion (5-10%), detected signals showed similar 'd' values (refer to Figure 34C). [0096] (B) A histogram describing the distribution of force. Average is 781 ± 151 pN (n=105) at a loading rate of 180 nN/sec.
[0097] (C) A definition of the distance, 'd', the z-directional moving distance of a piezoelectric device when DOPA-PEG molecule is fully stretched
[0098] (D) A plot of bonding strength (linear) vs. loading rate (log). Loading rate was the product of spring constant of a cantilever and a pulling speed. Four different loading rates were selected: 1500, 180.7, 28.4 and 2 nN/sec. Averaged forces with standard deviation were plotted at each given loading rate. Forces were 846.48 ± 157 pN (1500 nN/s), 781 ± 151 pN (180 nN/s), 744 ± 206 pN (28.4 nN/s), and 636.2 ± 150 (2 nN/s). [0099] (E) A schematic energy landscape of DOPA binding. External force tilted the landscape and lowered the energy barrier from the reaction coordinates. The slope (=kBT/xb) of the linear regression plot was 32.31 resulting in the distance to activation barrier (xb) was 1.27 A. The energy barrier height was determined by extrapolation when a loading rate equals to zero (Eb =28.1 kcal/mol).
Figure 35. Molecular identification of the adhesive origin of DOPA
[00100] Single molecule adhesion of tyrosine on TiO2 surfaces. No clear binding signals were detected (upper representative signal, n=639 out of 700 repeats) except the initial electrostatic interactions which were unavoidable in some cantilevers. Nonspecific adsorption signals (lower representative signal, n=61 out of 700). [00101] (B) Confirmation of tyrosine existence on the tip surface. Pi (π) electrons of tyrosine phenyl group specifically interact with gold π-electron.
[00102] (C) Force distributions of tyrosine binding to a gold surface. Tyrosine presented 398 ± 98 pN (180 nN/sec) adhesion strength.
Figure 36. The change of DOPA adhesiveness upon oxidation (DOPAquinone)
[00103] A schematic chemistry pathway of the formation and oxidation of DOPA.
DOPA was created by the action of tyrosine hydrxylase and subsequently oxidized to DOPAquinone by pH and the enzyme. It is unstable and reactive due to tendency of radical formation of DOPAquinone. It can crosslink with other DOPA molecules (di- DOPA) as well as reacts with amine groups from lysines. The arrows are the potential reactions found in other species not in mussel adhesive proteins.
[00104] (B) Representative force signals (n=16) with similar 'd' defined in Fig IC
(~ 50 nm). They were collected for 1 hr AFM experiment (1800 repeats) in a basic condition (20 mM Tris-Cl, pH 9.7). Time progress makes from the top to bottom of the graph. Red signals indicate DOPA-TiO2 and black signals for DOPAquinone-TiO2 [00105] (C) Force histograms after total analysis of 1800 F-D curves. The histogram at a low force region showed 178 ± 82 pN (n = 143) and the one at a high force region exhibited 741 ± 110 pN (n=51).
[00106] (D) A scatter plot of the number of events during a specified time window
(10 min). DOPA signals (circle, left y-axis) gradually decreased from twenty-two events for the first ten minutes to only three events during the last time window. However, quinone signals (triangle, right y-axis) increased from one event at the first time window to forty-two events at the last time window (50-60 min).
[00107] To summarize, successfully measured the single molecule binding strength of DOPA (-0.8 nN) was successfully measured, and the reversible binding chemistry was shown. This strong adhesion was created by post-translational modification but was significantly reduced by oxidation of DOPA, to DOPAquinone. [00108] The antifouling coating of the present invention can either be essentially permanent i.e., lasting 120 days or more, or biodegradeable depending on the number of DOPA or DOPA-derived moieties in the adhesive component. Figure 20 shows the results of a 28-day 3T3 fibroblast cell adhesion and spreading assay on TiO2 treated with mPEG-DOPA1-3. At early time points (i.e. less than 7 days), the protein and cell attachment resistance correlates well with the length of the DOPA peptide anchoring group, with resistance increasing in the order mPEG-DOPA < mPEG-DOPA2 < mPEG- DOPA3. TiO2 substrates treated with mPEG-DOPA2 and mPEG-DOPA3 maintain reduced cell attachment, or resistance, through 21 days. [00109] Robust design methodology was used to determine the effect of DOPA peptide anchor length and modification conditions (pH, concentration and time) on the PEG surface density and antifouling performance of metal, metal oxide, semiconductor, and polymer surfaces. The nine experiments utilized for each substrate are described below in Table 7. For nearly all surfaces, the length of the DOPA peptide and the pH of the modification buffer lead to the largest variation in the amount of PEG absorbed, as measured by XPS and the adhesion and spreading of 3T3 fibroblasts. The experiments summarized in Table 7 permitted the determination of modification conditions that provided optional cell adhesion resistance for a variety of materials, as measured by 4- hour cell attachment assays. After the nine experiments were performed on each substrate, the data was subjected to Robust Design analysis. The presence of large error values when plotting Robust Design data is a characteristic of the technique since a single data point at one factor level contains the variation of the remaining factors averaged over all the levels.
[00110] The polymeric compositions of the present invention can be also used to coat the surfaces of devices and instrumentation used for handling body fluids including sera. The coating on the surface of the device or instrument blocks protein binding to the surfaces thus reducing or eliminating the need for extensive washing or cleaning of the device or instrument between uses. The devices need to be thoroughly cleaned to prevent cross contamination between samples of bodily fluids applied top the device. Presently, the cleaning of these instruments and device between uses requires extensive washing with caustic agents such as 50% bleach and/or elevated temperatures. The coating process would be to circulate an aqueous solution of 1 mg/ml of DHPD polymer through the device at room temperature for a period of a few hours. [00111] The coatings of the present invention can be used on medical implants for a wide variety of uses. For example, the coatings can be used to block bacterial adhesion and therefore growth on the implanted device reducing the possibility of infection at the site of implant. The coatings can be used to reduce the amount of acute inflammation on the device by reducing protein binding and cell adhesion to the device. The coatings of the present invention can also be used as nanoparticles to prevent aggregation of these particle in the presence of serum. Hydrogels
[00112] The polymeric compositions of the present invention can also be as surgical adhesives for medical and dental uses and as vehicles for drug delivery to mucosal surfaces. The polymeric compositions can be used as tissue adhesive polymeric hydrogels for medical uses such as tissue sealants, gels for prevention of surgical adhesions (scar tissue formation), bone and cartilage adhesives, tissue engineering, and site specific drug elution and for research uses such as immobilization of proteins including antibodies and small molecule analytes including pharmaceuticals. The polymeric compositions of the present invention may also be used as interfacial bonding agents, wherein the neat monomers or solution of monomers are applied to a surface as a primer or bonding agent between a tissue surface or a metal or metal oxide implant/device surface and a bulk polymer or polymer hydro gel. With an appropriate polymer component, which one of ordinary skill in the art could identify, the polymeric compositions of the present invention can be injected or delivered in a fluid form and harden in situ to form a gel network. The in situ hardening can occur through photocuring, chemical oxidation, enzymatic reaction or through the natural increase in temperature resulting from delivery into the body.
[00113] In part, the present invention is also a method for the non-oxidative gelation of a polymeric composition of the present invention. One such method includes (1) providing a polymeric composition of the present invention; (2) admixing water and the polymeric composition; and (3) increasing admixture temperature sufficient to gel the polymeric composition, such temperature increase without oxidation of the polymer or DOPA or DOPA-derived moiety residue incorporated therein. Depending upon choice and identity of the polymer component of such a composition, an increase in admixture concentration can reduce the temperature required to effect gelation. Depending upon choice and identity of a particular copolymer component, a larger hydrophilic block thereof can increase the temperature required to gel the corresponding composition. Various other structural and/or physical parameters can be modified to tailor gelation, such modifications as can be extended to other polymeric compositions and/or systems which are consistent with the broader aspects of this invention.
[00114] It is widely acknowledged that the commercially-available PLURONIC® block copolymers self-assemble in a concentration- and temperature-dependent manner into micelles consisting of a hydrophobic PPO core and a water-swollen corona consisting of PEO segments. At high concentration, certain PEO-PPO-PEO block copolymers, such as PLURONIC® F127 and PLURONIC® F68, transform from a low viscosity solution to a clear thermoreversible gel at elevated temperature. While not wanting to be bound by theory, it is generally assumed that the interactions between micelles at elevated temperature lead to the formation of a gel phase, which is stabilized by micelle entanglements. The micellization and gelation processes depend on factors such as block copolymer molecular weight, relative block sizes, solvent composition, polymer concentration, and temperature. For example, increasing the length of the hydrophilic PEO blocks relative to the hydrophobic PPO block results in an increase in micellization and gelation temperature (Egeι).
[00115] Differential scanning calorimetry (DSC) measurements were performed on aqueous solutions of DME-PAO7 and DOPA-PAO7 at different concentrations to detect aggregation of block copolymers into micelles. DSC profiles obtained for
PLURONIC® F127, DME-PAO7 and DOPA-PAO7 were found to be qualitatively similar and were characterized by a large endothermic transition corresponding to micelle formation followed by a small endotherm at Tgs\ (Figure 2). The transition temperature of the small peak was found to correlate strongly with geι determined by rheometry and the vial inversion method (Table 4). Table 4 Gel temperatures obtained from vial inversion method, rheology or differential scanning calorimetry for 22 wt % DME-PAO7, DOPA-PAO7 and PLURONIC® F127 solutions. Gel Temperature (°C) vial inversion method Rheological DSC DME-PAO7 (22 wt %) 22.0 ± 1.0 20.3 ± 0.6 20.9 ± 0.1 DOPA-PAO7 (22 wt %) 22.0 ± 1.0 20.4 ± 0.5 21.7 ± 0.2 PLURONIC® F127 (22 wt 17.0 ± 1.0 15.4 ± 0.4 17.5 ± 0.4 %)
[00116] Aqueous solutions with concentrations ranging from 10 to 30 % (w/w) of
DOPA-PAO7 copolymers and 35 to 54 % (w/w) of DOPA-PAO8 copolymers were prepared by the cold method, in which DOPA conjugate was dissolved in distilled water at about 4°C with intermittent agitation until a clear solution was obtained. Thermal gelation of concentrated solutions was initially assessed using the vial inversion method. In this method, the temperature at which the solution no longer flows is taken as the gelation temperature.
[00117] The gelation temperature was found to be strongly dependent on copolymer concentration and block copolymer composition (i.e., PAO7 versus PAO8). For example, 22 wt % solutions of DOPA-P AO7 and DME-PAO8 were found to form a transparent gel at approximately 22.0 ± 1.0°C; decreasing the polymer concentration to 18 wt % resulted in a gelation temperature of approximately 31.0 ± 1.0°C. However, DOPA-PAO7 solutions with concentrations less than 17 wt % did not form gels when heated to 60°C. DOPA-PAO7 exhibits a slightly higher gel temperature than that (17.0 ± 1.0°C) of unmodified PLURONIC® F127. The gelation behavior of DOPA- PAO8 was found to be qualitatively similar, except that much higher polymer concentrations were required to form a gel. 54 wt % solutions of DOPA-PAO8 and DME-PAO8 formed gels at 23.0 ± 1.0°C, while 50 wt % of DOPA-PAO8 gels at 33.0 ± 1.0°C. However, DOPA-PAO8 solutions with concentrations less than 35 wt % did not form gels when heated to 60°C. DOPA-PAO8 exhibits a much higher gel temperature than that (16.0 ± 1.0°C) of unmodified PLURONIC® F68. These gels were found to be resistant to flow over long periods of time. From this experiment, we have also found that both DOPA and DOPA methyl ester-derivatives of the same commercially available PLURONIC® PAO exhibit almost the same gel temperature, and the gel made from 54 wt % of either DME-PAO8 or DOPA-PAO8 at room temperature is stiffer than that made from 22 wt % of either DME-PAO7 or DOPA-PAO7. [00118] The viscoelastic behavior of DOPA-modified PLURONIC® solutions was further studied by oscillatory rheometry. Figure 3 shows the elastic storage modulus, G', of 22 wt % solutions of unmodified PLURONIC® F127 and DME-PAO7 aqueous solutions as a function of temperature. Below the gelation temperature, storage modulus G ' was negligible, however G ' increased rapidly at the gel temperature (rgeι), defined as the onset of the increase of the G' vs. Temperature plot. DOPA-PAO7 (not shown) exhibited a similar rheological profile. The rgeι of 22 wt % solutions of DME- PAO7 and DOPA-PAO7 were found to be identical (20.3 ± 0.6°C), which is approximately 5 degrees higher than an equivalent concentration of unmodified- PLURONIC® F127 (15.4 ± 0.4°C). G' of DME-PAO7 or DOPA-PAO7 approaches a plateau value of 13 kPa, which is comparable to that of unmodified PLURONIC® F127. [00119] Shown in Figure 4 are the rheological profiles of 50 wt % solutions of unmodified PLURONIC® F68 and DME-PAO8 as a function of temperature. The Egeι of a 50 wt % DME-PAO8 solution was found to be 34.1 ± 0.6°e, whereas the rgeι of an equivalent concentration of unmodified PLURONIC® F68 was approximately 18°C lower (16.2 ± 0.8°C). The plateau storage moduli of 50 wt % solutions of DME-PAO8 and unmodified PLURONIC® F68 were not significantly different, approaching a plateau value as high as 50 kPa. The concentration dependence of Tgeι is illustrated in Figure 5, which shows the rheological profile of DME-PAO8 at two different concentrations as a function of temperature. Jgei of 45 wt % solution of DME-PAO8 was observed to be approximately 12°C higher than that of 50 wt % solution of DME-PAO8. [00120] Since both DOPA and DOPA methyl ester can be considered hydrophilic, the increase of rgeι observed in the DOPA-modified PLURONIC® PAOs, compared with that of unmodified PLURONIC® PAOs, is likely due to the increase in length of the hydrophilic PEO segments resulting from coupling of DOPA to the endgroups. It is clear from the data shown in Figures 3 and 4 that the coupling of DOPA or DOPA methyl ester to the PLURONIC® PAO endgroups has a more significant impact on the Tge\ of PLURONIC® F68 compared to PLURONIC® F127. This can be rationalized in terms of the overall molecular weights of F68 (approx. 8,600) and F127 (approx. 12,600). Addition of DOPA and DOPA methyl ester to both endgroups using the chemistry shown in Scheme 1 results in an increase in molecular weight of 446 and 474, respectively. This represents a larger % molecular weight increase for F68 compared to F127, due to lower base molecular weight of F68.
[00121] The data presented herein is in agreement with previous calorimetry studies of unmodified PLURONIC® PAOs, which demonstrated that the broad peak at low temperature is due to micellization while the small peak at higher temperature, only observed in concentrated solutions, corresponds to gelation, a nearly athermal process. As seen in Table 5, the onset temperature of micellization, the temperature at maximum heat capacity and rgeι of unmodified PLURONIC® F127 were found to be lower than those of DOPA-PAO7, whereas the specific enthalpies determined from the areas under the transition (Figure 2) are approximately the same. These enthalpies include contributions from both micellization and gelation. However, due to the small enthalpy of gelation, the observed enthalpy changes can be largely attributed to micellization.
Table 5
Comparisons of 30 wt % DME-PAO7, DOPA-PAO7 and unmodified PLURONIC® F127 solutions on onset micellization temperature, temperature at maximum heat capacity, enthalpies, and gel temperature from differential scanning calorimetry experiments Micellization Temp, at ΔH Geι Temp temp.(°C) Max. heat
Figure imgf000039_0001
„_ ' capacity (°C) DME-PAO7 (30 wt %) 5.2 ± 0.2 8.3 ± 0.1 20.3 ± 2.4 14.0 ± 0.4 DOPA-PAO7 (30 wt %) 4.6 ± 0.2 8.0 ± 0.6 19.3 ± 1.4 14.0 ± 0.2 PLURONIC® F127 (30 1.9 ± 0.3 6.0 ± 0.4 20.6 ± 1.6 10.6 ± 0.6 t %)
[00122] The micellization peak was seen to extend to temperatures above the onset of gelation, indicating that additional monomers aggregate into micelles at temperatures above the gelation point. The concentration dependence of DOPA-PAO7 and DME- PAO7 aggregation is shown in Figure 6. DSC thermograms indicate a decrease in micellization temperature and geι with increasing polymer concentration. The broad endothermic peak corresponding to micellization can also be observed in solutions at concentrations at which no gelation takes place; the characteristic temperature of the broad peak increases linearly with decreasing copolymer concentration, whereas the small peak was observed to coincide to the gel temperature of the concentrate copolymers but disappears as copolymer concentration decreases.
[00123] As can be gathered from the preceding, various polymeric compositions of this invention can be designed and prepared to provide various micellization and/or thermal gelation properties. Alternatively, or in conjunction therewith, degradation into excretable polymer components and metabolites can be achieved using, for instance, polyethylene glycol and lactic/glycolic acids, respectively. Regardless, the polymeric compositions of this invention provide improved adhesion by incorporation of one or more DHPD residues, such incorporation resulting from the coupling of a terminal monomer of the polymeric component to such a residue.
[00124] Another method of non-oxidative gelation of a polymeric composition of the present invention is photocuring. A photocurable DHPD-moiety-containing monomer is copolymerized with PEG-DA (PEG-diacrylate) to form adhesive hydrogels through photopolymerization. Photopolymerization can be achieved at any visible of UV wavelength depending on the monomer used. This is decidedly determined by one skilled in the art. The photocurable monomers consist of an adhesive moiety coupled to a polymerizable monomer with a vinyl group, such as a methacrylate group with or without an oligomeric ethylene oxide linker or fluorinated ether linker in between. [00125] An aqueous mixture of a photopolymerizable monomer containing an adhesive of the present invention and PEG-DA and 1.5 μL/mL of a photoinitiator, such as 2,2'-dimethoxy-2-phneyl-acetonephenone (DMPA), camphorquinone/4-
(dimethylamino)-benzoic acid (CQ/DMAB), and ascorbic acid/fluorescein sodium salt (AA/FNa2) was irradiated using a UV lamp (365 nm), for more than 5 minutes. Presence of the adhesive in the precursor solution was found to affect the radical polymerization process. The catechol adhesive decreased the extent of gel formation, reduced the percent of adhesive incorporation in the gel network, and lengthened the gel formation time.
[00126] As shown in Figure 25, gel conversion as determined by measuring the mass of the sondgel, reached more than 75 wt % after 2 minutes of UV irradiation and increased to greater than 85 wt % upon irradiation for more than 5 minutes. Gelation of PEG-DA occurred in 4 minutes or less when visible light initiators were used (4 minutes for CQ/DMAB and 3 minutes for AA/FNa2).
[00127] Copolymerization of PEG-DA with 1 or 7 (syntheses of 1 and 7 are shown in Schemes 2 and 3) was qualitatively similar to polymerization of pure PEG-DA, although addition of 1 or 7 to the PEG-DA precursor solution resulted in a decrease in gel conversion that was dependent on DOPA monomer concentration and initiating system. For example, in DMPA-initiated UV polymerization, gel conversion was reduced to less than 85 wt % in the presence of 2.5 mol % or more of 1 or 7. However, the extent of gel conversion was not statistically different between the gels. Similar DOPA concentration dependent inhibition was observed for the visible light induced initiators. For AA/FNa2 and CQ/DMAB initiated mixtures, addition of 33.3 mol % of 1 increased the gelation time to more than 8 minutes. Nevertheless, solutions containing 1 and 7 were still capable of photocuring even at a relatively high mol % of DOPA. SCHEME 2
Figure imgf000041_0001
DOPA
SCHEME 3
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000043_0002
[00128] For example, 53.8 mol % of either 1 or 7 in the precursor solution reduced the gel weight % from 88 to 77 and only 85 mol % of the DOPA was incorporated into the hydrogel. A colorimetric DOPA assay developed by Waite and Benedict performed on the photocured hydrogels revealed the presence of catecholic DOPA in the hydrogel. After photocuring, DOP A-containing gels were dialyzed in 0.5 N HCl to extract unreacted DOPA monomer. To quantify the extent of DOPA incorporation, the dialysate was analyzed according to the DOPA colorimetric assay of Waite and Benedict and the results were used to calculate the amount of DOPA incorporated into the gel network. Figure 26 shows the mole fraction of DOPA incorporated into the gel network, as a function of mol % monomer 1 and 7 in the precursor solution. There was no significant difference in the mole fraction of DOPA incorporated between samples containing 1 and 7. As much as 24.9 μmol/g of DOPA was incorporated into the PEG hydrogels. [00129] Direct evidence for the presence of DOPA in the gels was obtained by immersing the intact dialyzed hydrogels in nitrite reagent followed by NaOH. The initially colorless gels turned bright yellow after the addition of the nitrite reagent and then red following the addition of excess base. This color transition is typical of catechols, indicating that the unoxidized form of DOPA was incorporated into the hydrogels through photopolymerization. The intensity of the red color also reflects the concentration of DOPA incorporated into the photocured gels.
[00130] Contact mechanical tests were performed on the photocured gels in the shape of hemispheres to obtain information on the mechanical properties of the gels. Elastic moduli (E) were calculated by assuming Hertzian mechanics for the specific case of non-adhesive contact between an incompressible elastic hemisphere and a rigid plane, in which case the Hertzian relationship between load (P/,) and displacement (δh) becomes:
(1)
Figure imgf000044_0001
where R is the radius of curvature of the hemispherical gel. Load versus displacement data was fitted with Equation (1), which allowed the elastic moduli to be calculated based on the proportionality factor of the curve fit. As seen in Table 6, average Young's moduli (E) for DOP A-containing gel of around 50 kPa was obtained. Table 6 Average Young's Moduli for DOPA containing gels
Figure imgf000044_0002
33 mol % in the precursor solution * Determined from DOPA assay
** p< 0.001 relative to PEG-DA gels
[00131] These modulus values are about 30% lower than that of PEG-DA gels, confirming the inhibitory effect of DOPA on radical photopolymerization. Despite the decrease in modulus compared to PEG-DA gels, DOP A-containing gels still exhibited moduli suitable for many biomedical applications. A suitable modulus is one greater than 500 Pa. Another use of the adhesive hydrogels is for localized drug delivery. For example, adhesive hydrogels can be formed on a mucous membrane in the mouth or oral cavity. The hydrogels can be loaded with a drug such as an antibiotic and facilitate slow release of the drug over a period of time. They hydrogel can also be loaded with an analgesic and used to deliver pain relief at a localized site. They hydrogel can also be loaded with a chemotherapy drug and inserted into malignant tissue to deliver localized cancer therapy. The hydrogel can also be loaded with a cell proliferation inhibitor therapeutic drug and used as a coating stent or other vascular device and used to control cell proliferation at the site of an implant of the vascular device.
[00132] A tissue adhesive hydrogel capable of being cross-linked in vivo can be used as a tissue sealant for replacing metal or plastic sutures. The adhesive bends to the surrounding tissue at a surgical or injury site and the polymer forms a cohesive link to close the wound. The hydrogel can also be used for repair of bone fractures and cartilage to bone damage.
Other Uses
[00133] There are various industrial product uses of these coatings and hydrogels including prevention of marine biofouling (attachment of algae, bacteria, and mussels to surfaces underwater), prevention of bacteria contamination of water streams to industrial plants such as electronic and drug manufacturers, prevention of bacterial contamination of drinking water streams, dental and denture adhesives, underwater adhesives to deliver indicators, coatings for water purity and measurement sensors, paints used for prevention of biofouling.
[00134] There are also a number of consumer product and cosmetic uses of these coatings and hydrogels including without limitation use in dental and denture adhesives, use in cosmetics for adhesive to hair, skin, and legs, use in cosmetics such as eye shadow, lip stick, and mascara, use in application of temporary tattoos, and use as resealable adhesives for bags and containers.
[00135] Without limitation to any particular synthetic scheme or method of preparation, suitable compositions of this invention can include but are not limited to a urethane moiety between each such terminal monomer and DOPA residue. As described more fully below, such a moiety is a synthetic artifact of the agent/reagent utilized to couple the DOPA residue with the polymeric component. Within the broad aspects of this invention, various other moieties are contemplated, as would be understood by those skilled in the art made aware of this invention, depending upon terminal monomer functionality and choice of coupling agent. Examples: General Description [00136] The following non-limiting examples and data illustrate various aspects and features relating to the compositions and/or methods of the present invention, including the production of various polymeric or co-polymeric compositions having incorporated therein one or more DHPD components, as are available through the synthetic methodology described herein. While the utility of this invention is illustrated through the use of several polymeric or co-polymeric systems, it will be understood by those skilled in the art that comparable results are obtainable with various other compositions and/or methods for preparation, as are commensurate with the scope of this invention.
[00137] PEOiooPPO65PEOioo (PLURONIC® F127, avg. Mw = 12,600),
PEO78PPO30PEO78 (PLURONIC® F68, avg. Mw = 8,400), PEG (avg. Mw = 8000), pentafluorophenol, 1,3-dicyclohexylcarbodiimide (DCC), 4,7, 10-trioxa- 1,13- tridecanediamine, fluorescein sodium salt (FNa2), and ascorbic acid (AA) were purchased from Sigma (St. Louis, MO). L-DOPA, thionyl chloride, methacroyloyl chloride, /-butyldimethylsilyl chloride (TBDMS-C1), di-t-butyl dicarbonate, methacrylic anhydride, 2,2'-dimethoxy-2-phenyl-acetonephenone (DMPA), acryloyl chloride, 1,8- diazabicyclo[5.4.0]undec-7-ene (DBU), tetrabutylammonium fluoride (TBAF), 4- (dimethylamino)-benzoic acid (DMAB), l-vinyl-2 -pyrrolidone (VP), N,N-disuccinimidyl carbonate, sodium borate, sodium molybdate dihydrate, sodium nitrite, 4- (dimethylamino)pyridine (DMAP), Ν-hydroxysuccinimide, Ν,Ν-diisopropylethylamine, dimethylformamide, and dichloromethane were purchased from Aldrich (Milwaukee, WI). Camphorquinone (CQ) was obtained from Polysciences, Inc. (Warrington, PA). Acetone was dried over 4A molecular sieve and distilled over P2O5 prior to use. Triethylamine was freshly distilled prior to use. All other chemical reagents were used as received.
[00138] L-DOPA methyl ester hydrochloride was prepared according to the procedure of Patel and Price, J. Org. Chem., 1965, 30, 3575, which is incorporated herein by reference. Succinimidyl propionate activated PEG (mPEG-SPA, avg. Mw = 5000) was obtained from Shearwater Polymers, Inc. (Huntsville, AL). Ethyl acetate saturated with HCl was prepared by bubbling HCl gas through ethyl acetate (50 mL) for approximately 10 minutes. 3,4-Bis(t-butyldimethylsiloxyl)L-phenylalanine (DOP A(TBDMS)2) and 3 ,4-bis(t-butyldimethylsiloxyl-N-t-butoxycarbonyl-L- phenylalanine (Boc-DOPA(TBDMS)2) were synthesized according to the method of Sever and Wilker, Tetrahedron, 2001, 57, (29), 6139-6146, which is incorporated herein by reference.
[00139] Glass coverslips (12mm dia.) used in the following examples were cleaned by immersing in 5% Contrad70 solution, a detergent which is an emulsion of anionic and nonionic surfactants in an allealtine aqueous base (Decon Labs, Inc., Bryn Mawr, PA) in an ultrasonic bath for 20 minutes, rinsed with deionized (DI) H2O, sonicated in DI H2O for 20 minutes, rinsed in acetone, sonicated in acetone for 20 minutes, rinsed in hexanes, sonicated in hexanes for 20 minutes, rinsed in acetone, sonicated in acetone for 20 minutes, rinsed in DI H2O, and sonicated in DI H2O for 20 minutes. The coverslips were subsequently air-dried in a HEPA-filtered laminar flow hood. To create pristine gold substrates, clean coverslips were sputtered (Cressington 208HR) with 2 nm Cr followed by 10 mn Au (99.9% pure).
[00140] Titanium oxide (TiO2) surfaces were prepared by electron beam physical evaporation onto silicon (Si) wafer and cleaned in a plasma chamber prior to testing. Si wafers (MEMC Electronic Materials, St. Peters, MO, surface orientation (100)) were coated with 100 mn Ti by an Edwards FL400 electron beam evaporator at <10" Torr. The Si wafer was then cut into 8mm x 8mm pieces which were subsequently cleaned by ultrasonication in the following media: 5% Contrad70, ultrapure water (ultrapure water is deionized and distilled), acetone, and petroleum ether. The substrates were further cleaned in an oxygen plasma chamber (Harrick Scientific, Ossining, NY) at <200 mTorr and 100W for 3 minutes.
[00141] Both pristine and modified gold surfaces were characterized, as described below, by X-ray photoelectron spectroscopy (XPS). XPS data was collected on an Omicron ESCALAB (Omicron, Taunusstein, Germany) configured with a monochromated AlKα (1486.8eV) 300-W X-ray source, 1.5mm circular spot size, a flood gun to counter charging effects, and an ultrahigh vacuum (<10"8 Torr). The takeoff angle, defined as the angle between the substrate normal and the detector, was fixed at 45°. Substrates were mounted on standard sample studs by means of double-sided adhesive tapes. All binding energies were calibrated using either the Au(4/7/2) gold peak (84.0eV) or the C(\s) carbon peak (284.6eV). Analysis consisted of a broad survey scan (50.0eV pass energy) and a 10-minute high-resolution scan (22.0eV pass energy) at 270-300eV for C(l^). Peak deconvolution and atomic percent calculations were performed with EIS analysis software.
[00142] Secondary ion spectra were collected on a TRIFT III™ time-of flight secondary ion mass spectrometer (TOF-SIMS) (Physical Electronics, Eden Prairie, MN) in the mass range 0-2000 m/z. A Ga+-source was used at a beam energy of 15 keV with a 100 μm raster size. Both positive and negative spectra were collected and calibrated with a single set of low mass ions using the PHI software Cadence.
[00143] To determine relative hydrophilic/hydrophobic nature of the surfaces, contact angle data was collected, as described below, by the sessile drop method. A custom-built contact angle goniometer (components from Rame-Hart, Mountain Lakes, NJ) equipped with a humidified sample chamber was used to measure both advancing and receding contact angles of ultrapure water (18.2MΩ-cm; Barnstead, Dubuque, IA) on unmodified and modified substrates. For each surface, four measurements were made at different locations and the mean and standard deviation were reported. [00144] Surface Plasmon Resonance (SPR) measurements were made on a
BIACORE 2000 (Biacore International AB; Uppsala, Sweden) using bare gold sensor cartridges. The resonance response was calibrated using 0-lOOmg/ml NaCl solutions. Dilute solutions (0.1 mM in H2O) of mPEG-DOPA, mPEG-MAPd, and mPEG-OH were injected into the SPR flow cell for 10 minutes after which flow was switched back to pure DI H O. In a separate experiment to measure protein adsorption to modified substrates, sensor surfaces with preformed PEG films were exposed to O.lmg/ml bovine serum albumin (BSA) solution in 10 mM HEPES buffer (0.15 M NaCl, pH=7.2), and subsequently pure buffer.
[00145] For use in demonstration of anti-fouling effects, NIH 3T3-Swiss albino fibroblasts obtained from ATCC (Manassas, VA) were maintained at 37°C and 10% CO in Dulbecco's modified Eagle's medium (DMEM; Cellgro, Herndon, VA) containing 10% (v/v) fetal bovine serum (FBS) and lOOU/ml of both penicillin and streptomycin. [00146] RP-HPLC preparation was performed using a Waters HPLC system
(Waters, Milford, MA) on a Vydac 218TP reverse phase column with a gradient of acetonitrile /0.1% trifluoroacetic acid (v/v) water. ESI-MS analysis was performed on a LCQ LC-MS system (Finnigan, Thermoquest, CA). MALDI-TOF MS analysis was performed on a Voyager DE-Pro mass spectrometer (Perseptive Biosystem, MA), α- Cyano-4-hydroxycinnaminic acid was used as a matrix. NiTi alloy (10 mm x 10 mm x 1 mm) was obtained from Nitinol Devices & Components (Fremont, CA). Si, SiO2 (1500A thermal oxide), and GaAs wafers were purchased from University Wafer (South Boston, MA).
[00147] With regard to the following cell adhesion tests and/or spreading assays, modified and unmodified substrates were pretreated in 12-well TCPS plates with 1.0 ml of DMEM containing 10% FBS for 30 minutes at 37°C and 10% CO2. Fibroblasts of passage 12-16 were harvested using 0.25% trypsin-EDTA, resuspended in DMEM with 10% FBS, and counted using a hemocytometer. Cells were seeded at a density of 2.9 x 103 cell/cm2 by diluting the suspension to the appropriate volume and adding 1 ml to each well. The substrates were maintained in DMEM with 10% FBS at 37°C and 10% CO2 for 4 hours, after which time unattached cells were aspirated. Adherent cells on the substrates were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently treated with 5 μM l,l'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil; Molecular Probes, Eugene, OR) in DMSO for 30 minutes at 37°C. The stain was then aspirated and substrates were washed (3x) with DMSO for 10 minutes and mounted on glass slides using Cytoseal (Stephens Scientific, Kalamazoo, MI) to preserve fluorescence. These experiments were performed in triplicate for statistical purposes. For electron microscopy, some samples were dehydrated with EtOH after fixing, critical- point dried, and sputtered with 3 nm Au.
[00148] To quantify cell attachment, substrates were examined with an Olympus
BX-40 (λπx = 549nm, λEm = 565nm) and color images were captured with a Coolsnap CCD camera (Roper Scientific, Trenton, NJ). Five images were taken from each of the three substrate-replicates. The resulting images were quantified using thresholding in MetaMorph (Universal Imaging, Downington, PA). A one-way ANOVA and Tukey's post-hoc test with 95% confidence intervals (SPSS, Chicago, IL) were used to determine statistical significance of the data. The mean and standard deviation of the measurements were reported. Example 1
Synthesis of Succinimidyl Carbonate PAO, SC-PA07
[00149] PLURONIC® F127 (0.60 mmols) was dissolved in 30 mL of dry dioxane.
N,NJ-Disuccinimidyl carbonate (6.0 mmols) in 10 mL dry acetone was added. DMAP (6.0 mmols) was dissolved in 10 mL dry acetone and added slowly under magnetic stirring. Activation proceeded 6 hours at room temperature, after which SC-PAO7 was precipitated into ether. The disappearance of the starting materials during the reaction was followed by TLC in chi oroform-m ethanol (5:1) solvent system. The product was purified by dissolution in acetone and precipitation with ether, four times. The product yield was 65%. 1H ΝMR (500 MHz, CDC13): δ ppm 0.96-1.68 (br, -OCHCH3CH2O-), 2.80 (s, -COON(CO)2(CH2)2), 3.15-4.01 (br, -OCH2CH2O-; -OCHCΗ3CH2O-), 4.40 (s, -OCΗ2CH2OCOON(CO)222-). Example 2 Synthesis ofDME-PA07
[00150] A slurry of DOPA methyl ester hydrochloride (1.25 mmols) and triethylamine (2.5 mmols) was mixed with SC-PAO7 (0.16 mmols) in 10 mL chloroform. The disappearance of the starting materials during the reaction was followed by TLC in chloroform-methanol-acetic acid (5:3:1) solvent system. After stirring for 1 hour at room temperature, the solvent was evaporated off, and DME-PAO7 was purified by precipitation from cold methanol three times. DME-PAO7 gave a positive Arnow test indicating the presence of catechol hydroxyl groups. The product yield was 75%. 1H NMR (500 MHz, CDC13): δ ppm 0.98-1.71 (br, -OCHCH3CH2O-), 2.83-3.06 (m, -NHCHCH2C6H3(OH)2COOCH3), 3.15-4.02 (br, -OCH2CH2O-; ' -NΗCΗ(CΗ2C6Η3(OΗ)2COOCΗ3), 4.05-4.35 (d, -OCH2CH2OCONHCHCH2C6H3(OH)2COOCH3), 4.55
(br, -NHCHCH2C6H3(OH)2COOCH3), 5.30 (d, -NHCHCH2C6H3(OH)2COOCH3), 6.45-6.80 (Is, 2d, -NHCHCH2C6H3(OH)2COOCH3). Example 3 Synthesis ofDOPA-PA07
[00151] L-DOPA (1.56 mmols) was added to 30 mL 0.1 M Na2B4O7 (pH = 9.32) aqueous solution under Ar atmosphere, followed by stirring at room temperature for 30 minutes. SC-PAO7 (0.156 mmols) in 5 mL acetone was added to the resulting mixture and stirred overnight at room temperature. The solution pH was maintained with sodium carbonate during the reaction. The disappearance of the starting materials during the reaction was followed by TLC in chloroform-methanol-acetic acid (5:3:1) solvent system. The solution was acidified to pH 2 with concentrated hydrochloric acid and then extracted three times with dichloromethane. The combined dichloromethane extracts were dried with anhydrous sodium sulfate and filtered, and dichloromethane was evaporated. The product was further purified by precipitation from cold methanol. DOPA-P AO7 gave a positive Arnow test indicating the presence of catechol hydroxyl groups. The product yield was 52%. 1H NMR (500 MHz, CDC13): δ ppm 0.92-1.70 (br, -OCHCH3CH2O-), 2.91-3.15 (m, -NHCHCH2C6H3(OH)2COOCH), 3.20-4.10 (br, -OCH2CH2O-; -OCHCΗ3CH2O-), 4.1-4.35 (d, -OCΗ2CH2OCONΗCΗCΗ2C6Η3(OΗ)2COOΗ), 4.56
(m, -NHCHCH2C6H3(OH)2COOH), 5.41 (d, -NHCHCH2C6H5(OH)2COOH), 6.60-6.82 (Is, 2d, -NHCHCH2C6H3(OH)2COOH). Example 4 Synthesis of Succinimidyl Carbonate PA08, SC-PA08
[00152] A procedure similar to that described above for the synthesis and purification of SC-PAO7 was used to prepare SC-PAO8. The product yield was 68%. 1H NMR (500 MHz, CDC13): δ ppm 0.95-1.58 (br, -OCHCH3CH2O-), 2.80 (s, -COON(CO)2(CH2)2), 3.10-4.03 (br, -OCH2CH2O-; -OCHCΗ3CH2O-), 4.40 (s, -OCΗ2CH2OCOON(CO)222). Example 5 Synthesis ofDME-PA08
[00153] A procedure similar to that described above for the synthesis and purification of DME-PAO7 conjugate was used to make DME-PAO8. The product yield was 76%. 1H NMR (500 MHz, CDC13): δ ppm 0.98-1.50 (br, -OCHCH3CH2O-), 2.85-3.10 (m, -NHCHCH2C6H3(OH)2COOCH3), 3.15-4.01 (br, -OCH2CH2O-; -OCHCΗ3CH2O-; -NΗCΗ(CΗ2C6Η3(OΗ)2COOCH3), 4.03-4.26 (d, -OCΗ2CH2OCONΗCΗCΗ2C6Η3(OΗ)2COOCΗ3), 4.55
(m, -NHCHCH2C6H3(OH)2COOCH3), 5.30 (d, -NHCHCH2C6H3(OH)2COOCH3), 6.45-6.77 (Is, 2d, -NHCHCH2C6H3(OH)2COOCH3). Example 6
Synthesis of DOPA-P A08
[00154] A procedure similar to that described above for the synthesis of DOP A-
PAO7 conjugate was used to prepare and purify DOPA-PAO8. The product yield was
49%. 1H NMR (500 MHz, CDC13): δ ppm 0.92-1.50 (br, -OCHCH3CH2O-), 2.91-3.10
(m, -NHCHCH2C6H3(OH)2COOH), 3.15-3.95 (br, -OCH2CH2O-; -OCHCΗ3CH2O-),
4.06-4.30 (d, -OCΗ2CH2OCO NΗCΗCΗ2C6Η3(OΗ)2COOΗ), 4.54
(m, -NHCHCH2C6H3(OH)2COOH), 5.35 (d, -NHCHCH2C6H5(OH)2COOH), 6.50-6.80
(1 s, 2d, -NHCHCH2C6H3(OH)2COOH). Example 7 Colorimetric Assay
[00155] Coupling efficiencies of DOPA methyl ester and DOPA to
PLURONICs® F127 and F68 were determined using the colorimetric method of Waite and Benedict. Briefly, samples were analyzed in triplicate by diluting aliquots of standards or unknown solutions with 1 N HCl to a final volume of 0.9 mL. 0.9 mL of nitrite reagent (1.45 M sodium nitrite and 0.41 M sodium molybdate dihydrate) was added to the DOPA solution, followed immediately by the addition of 1.2 mL of 1 N NaOH. Due to time-dependent changes in absorbance intensity, care was taken to ensure that the time between the addition of NaOH and recording of the absorbance was 3 minutes for all standards and samples. The absorbance was recorded at 500 nm for all standards and samples. DOPA was used as the standard for both the DOPA methyl ester and DOPA conjugates. Example 8 Rheology
[00156] Rheological measurements of the gelation process were performed using a
Bohlin VOR Rheometer (Bohlin Rheologi, Cranbury, NJ). A 30 mm diameter stainless steel cone and plate geometry with a cone angle of 2.5 degrees was used for all measurements. The temperature was controlled by a circulating water bath. Samples were cooled in the refrigerator prior to transfer of 0.5 mL of liquid solution to the apparatus. Measurements of storage and loss moduli, G' and G", were taken in the oscillatory mode at 0.1 Hz and a strain of 0.45%. The heating rate was 0.5°C/min except in the vicinity of the gelation temperature, when it was reduced to 0.1°C/min. The strain amplitude dependence of the viscoelastic data was checked for several samples, and measurements were only performed in the linear range where moduli were independent of strain amplitude. Mineral oil was applied to a ring surrounding the outer surfaces of the sample compartment to prevent dehydration during measurements. Example 9 Differential Scanning Calorimetry (DSC)
[00157] DSC measurements were performed on a TA Instruments DSC-2920 (TA
Instruments, New Castle, DE) calorimeter. Spectra were obtained for three samples of each concentration on heating and cooling cycle. Sample volumes of 20 μl in hermetically sealed aluminum pans were used and scans were recorded at a heating and cooling rate of 3°C/min with an empty pan as reference. Example 10a [00158] Amino-terminated methoxy-PEG, mPEG-NH2 (2.0 g, 0.40 mmoles, Mw =
2,000 or 5,000, Sun-Bio PEGShop), N-Boc-L-DOPA dicyclohexylammonium salt (0.80 mmoles), HOBt (1.3 mmoles), and Et3Ν (1.3 mmoles) were dissolved in 20 mL of a 50:50 mixture of dichloromethane (DCM) and DMF. HBTU (0.80 mmoles) in 10 mL of DCM was then added, and the reaction was carried out under argon at room temperature for 30 minutes. The reaction solution was successively washed with saturated sodium chloride solution, 5% NaHCO3, diluted HCl solution, and distilled water. The crude product was concentrated under reduced pressure and purified by column chromatography on Sephadex® LH-20 with methanol as the mobile phase. The product, mPEG-DOPA, was further purified by precipitation in cold methanol three times, dried in vacuum at room temperature, and stored under nitrogen at -20°C. 1H NMR (500 MHz, CDC13/TMS): δ 6.81-6.60 (m, 3H, C6H3(OH)2-), 6.01 (br, s, IH, OH-), 5.32 (br, s, IH, OH-), 4.22 (br, s, IH, C6H3(OH)2-CH2-CH(N-)-C(O)N-), 3.73-3.38 (m, PEO), 3.07 (m, 2Η, PEO-CH2-NH-C(O», 2.73 (t, 2H, C6H3(OH)2-CH2-CH(N-)-C(O)N-), 1.44 (s, 9 H, (CH3)3C-), 1.25 (s, 3 H, CH3CH2O-).
Figure imgf000054_0001
Example 10b
[00159] The synthesis and related procedures of the preceding example can be extended, by analogy, using other DOP A-containing peptides and oligopeptides, whether natural or synthetic in origin. Depending upon a particular synthetic sequence, use of an N-terminal protecting group may be optional. As referenced above, various other DOPA-like adhesive components can also be utilized, as would be well-known to those skilled in the art made aware of this invention. For instance, B -amino acids and N-substituted glycine DOPA analogs can be used.
[00160] Regardless of a particular DHPD adhesive component, a variety of polymeric components can be used in accordance with the synthetic techniques and procedures described above. The polymeric component can vary in molecular weight limited only by corresponding solubility concerns. As mentioned above, a variety of other polymers can be used for surface anti-fouling and/or particle stabilization, such polymers including but not limited to hyaluronic acid, dextrans and the like. Depending upon solubility requirements and desired surface effect, the polymeric component can be branched, hyperbranched or dendrimeric, such components available either commercially or by well-known synthetic techniques.
[00161] While the composition of Example 10a is the amidation product of the referenced starting materials, it should be understood that comparable polymer-DHPD conjugates can be prepared coupling the N-terminus of a DHPD component to an end group, back bone or side chain of a suitably functionalized natural or synthetic polymer, including those described above. For example, and without limitation, as illustrated above, a suitable polymeric component terminating with a carbonate functionality can be used to provide the desired conjugate by reaction with the N-terminus of the desired DHPD component. Example 11a [00162] The consensus decapeptide repeat sequence (mussel adhesive protein decapeptide, MAPd, NH2-Ala-Lys-Pro-Ser-Tyr-Hyp-Thr-DOPA-Lys-CO2H) of the blue mussel Mytilus edulis foot protein 1 (Mefp 1) was synthesized by solid phase peptide synthesis on Rink resin (0.6 mMol/g) using Fmoc protected amino acids, BOP, HOBt, and DIEA as activating agents, and NMP as solvent. Fmoc deprotection was performed using a 25% piperidine solution in NMP for twenty minutes. Couplings of amino acids were performed using two equivalents of the Fmoc-amino acid BOP:HOBt:DIEA in a 1:1:1:1 ratio for twenty minutes, with an initial, ten-minute preactivation step. Upon completion of the decapeptide, the free amine terminus of the decapeptide was coupled to activated methoxy-PEG-CO2H (mPEG-SPA, Mw = 2k or 5k, Shearwater Polymers) using carbodiimide chemistry. The PEG-decapeptide conjugates (mPEG-MAPd, 2k or 5k) were cleaved at 0°C for two hours using 1 M TMSBr in TFA, with EDT, thioanisole, and m-cresol. The crude mPEG-MAPd products were precipitated in ether at 0°C, and purified by preparative HPLC using a Vydac 218TP reverse phase column (220x 22mm x lOμm). The purity of the products was determined to be >90% using analytical HPLC, and the structures confirmed using a PerSeptive Biosystem MALDI-TOF-MS. Example lib [00163] The synthesis and procedures of Example 11a can be extended analogous to and consistent with the variations illustrated in Example 10b. In addition, other conjugates can be prepared using DOP A-containing polymers prepared by enzymatic conversion of tyrosine residues therein. Other techniques well-known in the field of peptide synthesis can be used with good effect to provide other desired protein sequences, peptide conjugates and resulting adhesive/anti-fouling effects. Example 12a [00164] Gold surfaces were modified by adsorption of mPEG-DOPA or mPEG-
MAPd (2k, 5k) from solution in DCM or phosphate-buffered saline (PBS; pH = 3, 7.4, and 11) at polymer concentrations ranging from 0.1-75 mg/ml. Substrates were placed in a vial and immersed in mPEG-DOPA or mPEG-MAPd solution for up to 24 hours without agitation. Upon removal from solution, substrates were rinsed with the appropriate solvent (DCM or DI H2O) to remove unbound polymer, and dried in vacuo. For comparison, identical surface modifications were performed using PEG- monomethylether (mPEG-OH, avg. Mw = 5000). Alternatively, a drop of solution containing mPEG-DOPA or mPEG-MAPd (10 mM in PBS, PEG molecular weight=2000) was incubated on a Au-coated glass coverslip (Au thickness —10 nm) for 30 minutes at 37°C, after which the surface of the coverslip was rinsed (3x) with PBS. Analysis of the modified surfaces by advancing/receding contact angle, XPS, and TOF- SIMS revealed the formation of a chemisorbed layer of mPEG-DOPA or mPEG-MAPd. [00165] Figures 7A-C shows the XPS spectra for the unmodified, mPEG-OH modified, and mPEG-DOPA modified surfaces. As expected, the ether peak at 286.5 eV increased only slightly with the mPEG-OH treatment, while a dramatic increase was observed after adsorption of mPEG-DOPA, indicating a large presence of ether carbons. An ether peak from a pure PEG with the same binding energy has been reported in the literature. The smaller peak at 285.0 eV in Figure 7 can be attributed to the aliphatic and aromatic carbons in the PEG and DOPA headgroup, as well as some hydrocarbon contamination resulting from the preparation/evacuation process.
[00166] Time-of-flight SIMS data corroborated the XPS findings. TOF-SIMS analysis was carried out on unmodified and mPEG-DOPA-modified Au substrates, as well as mPEG-DOPA powder and a gold substrate exposed to mPEG-OH. Data was collected from each substrate for about 4 minutes.
[00167] The positive ion spectrum of unmodified Au exhibits (CnH2n+ι)+ ' and
(CnH2n-ι)+ peaks, typical for hydrocarbon contamination (data not shown). Additional minor contaminants were present, including NH4 +, Na+, and relatively small amounts of CaHbO c + species. Because of the process used to deposit the Au film, a peak for Cr was seen at m/z - 52, in addition to the Au peak at m/z - 196.9. Exposing the gold surfaces to mPEG-OH resulted in only modest increases in the peaks representing CaHbOc + PEG fragments, which are likely attributable to contamination or non-specific absorption of mPEG-OH. This is evidenced by the peaks at m/z - 225 (AuOC+) and 254 (AuOCCO+) which did not show dramatic increases when compared to substrates modified with mPEG-DOPA. (Figures 8A-C).
[00168] The positive ion spectrum of the Au surface modified with mPEG-DOPA was dominated by the presence of CaHbOc + peaks representing the adsorbed molecule. As illustrated in Figure 9, the relative abundance of C2H3O and C2H5O increased with respect to unmodified and mPEG-OH modified surfaces. There was also a dramatic increase in the relative abundance of C3H7 (m/z — 43) and C4H5 + (m/z — 53), as well, which can likely be attributed to hydrocarbon contamination or the fragmentation of the t-butyl in the Boc protection group.
[00169] Perhaps the most notable feature of the positive ion spectrum of the
PEGylated Au substrate were the patterned triplet repeats in the high mass range (Figure 10). Each of these triplet clusters corresponds to an Au-DOPA-(CH2CH2O)n fragment. When further resolved, each subcluster within the triplet represents the addition of CH2, CH2CH2, or CH2CH2O, as each of these peaks is about 14-16 amu apart. This repeat pattern was identifiable from n = 0-15, beyond which the signal was below detectable limits.
[00170] In the negative ion spectra for the pristine Au surface, little of note was observed aside from the strong definable peaks for O", HO", and Aun " for n = 1-3 (data not shown). There was a small amount of hydrocarbon contamination present at m/z - 13 (CH"), 24 (C2H2 "), and 37 (C3H"). The negative ion spectrum of the PEGylated Au surface was dominated by the peak for C HπO + at m/z - 126.893. The presence of this peak at modest intensity in the spectrum of the mPEG-OH modified Au suggests that it represents a larger ethylene glycol fragment. The most interesting peaks lie in the high mass range (>200 m/z) and represent the coupling of catecholic oxygen to Au. The spectrum suggests that one Au atom can bind up to six oxygen atoms, corresponding to three DOP As.
[00171] The contact angle data demonstrated a firm dependence on the character of the adsorption solvent used when modifying the gold films with mPEG-DOPA (data not shown). The surface modified in DCM showed a significantly lower θa than the unmodified surface (p<0.001) and the surfaces modified in all aqueous solutions (p<0.05). Generally speaking, as the pH of the aqueous solutions was increased, the hydrophilicity of the treated surfaces was decreased, indicating a diminished ability to PEGylate the surfaces, perhaps due to the propensity of DOPA to be oxidized to its less adhesive quinone form at elevated pH, an interpretation that is supported by previous studies that showed the unoxidized catechol form of DOPA is primarily responsible for adhesion. Example 12b
[00172] Protein adsorption and attachment/spreading of cells onto untreated and treated coverslips were evaluated as follows. Surface plasmon resonance (SPR) experiments demonstrated that the DOP A-containing polymers were rapidly bound to the gold surface and the resulting modified surfaces possessed an enhanced resistance to protein adsorption (Figure 11). Protein adsorption onto mPEG-MAPd (5k) modified gold was roughly 70% less than to the unmodified gold surface. Analysis of fibroblasts cultured on modified substrates showed a strong dependence of cell attachment on mPEG-DOPA concentration (Figure 12), adsorption solvent, and modification time used during preparation of the PEG-modified substrates. Surfaces modified for 24 hours with >25 mg/ml mPEG-DOPA or mPEG-MAPd exhibited a statistically significant reduction in cell attachment and spreading (Figures 12-14). The mPEG-MAPd (5k) modified gold surface exhibited a 97% reduction in total projected cellular area and a 91% reduction in the density of cells attached to the surface. Example 12c [00173] The modification illustrated in Example 12a, optionally varied as referenced in Examples 10b and l ib, can be extended to other noble metals, including without limitation, silver and platinum surfaces. Such application can also be extended, as described herein, to include surface modification of any bulk metal or metal alloy having a passivating or oxide surface. For example, bulk metal oxide and related ceramic surfaces can be modified, as described herein. Such techniques can also be extended to semiconductor surfaces, such as those used in the fabrication of integrated circuits and MEMS devices, as also illustrated below in the context of nanoparticulate stabilization. Example 13 [00174] Silicate glass surfaces (glass coverslips) were modified by adsorption of mPEG-MAPd (2k) from a 10 mM solution in water, using the method described in Example 12a. The cell density of NIH 3T3 cells attached to modified and unmodified glass surfaces were evaluated as described, above. Glass surfaces modified for 24 hours with mPEG-MAPd exhibited a 43% reduction in cell density compared to unmodified glass surfaces (Cell Density (cells/mm2): 75.5 +/- 6.5 on unmodified glass; 42.7 +/- 9.8 on mPEG-MAPd modified glass). Example 14a
[00175] To illustrate stabilization of metal oxides and, in particular, metal oxide nanoparticles, 50 mg of mPEG-DOPA (5k) was dissolved in water (18 MΩ-cm, Millipore) and combined with 1 mg of magnetite (Fe3O4) powder. Similar preparations were also prepared using a mPEG-NH2 (5k) (Fluka) and a mPEG-OH (2k) (Sigma) as controls. Each of these aqueous solutions was sonicated using a Branson Ultrasonics 450 Probe Sonicator for one hour while being immersed in a 25°C bath. The probe had a frequency of 20 kHz, length of 160 mm, and tip diameter of 4.5 mm. The sample was then removed and allowed to stand at room temperature overnight to allow any unmodified magnetite to precipitate out of solution. Suspensions prepared using the control polymers (mPEG-NH2 and mPEG-OH) rapidly precipitated to yield a brown solid and clear, colorless supernatant. In samples prepared using PEG-DOPA stabilized nanoparticles, the sample was clear and brown. The clear brown supernatant was isolated and dialyzed for three days in water using Spectra/Por® membrane tubing (MWCO: 15,000). Following dialysis, the sample was lyophilized and stored under vacuum at room temperature until used. Example 14b [00176] mPEG-DOPA stabilized nanoparticles were characterized by transmission electron microscopy (TEM), thermogravimetric analysis (TGA), fourier transform infrared spectroscopy (FTIR), and UV/vis spectroscopy. TEM results demonstrated that the majority of nanoparticles were of diameter of 5-20 nm (data not shown). TGA analysis of 0.4 mg of mPEG-DOPA stabilized magnetite indicated that the particles contain 17% by weight mPEG-DOPA (data not shown). FTIR performed on untreated magnetite showed relatively little absorbance within the wavelength range from 4000- 400 cm"1, whereas the mPEG-DOPA treated nanoparticles exhibited absorption bands at 800-1600 cm"1 and 2600-3200 cm"1, confirming the presence of mPEG-DOPA. Example 14c [00177] The dry PEG-DOPA stabilized magnetite nanoparticles readily dispersed in aqueous and polar organic solvents (e.g., dichloromethane) to yield clear brown suspensions that were stable for months without the formation of noticeable precipitates. Suspensions of mPEG-DOPA stabilized nanoparticles in various solvents were prepared by dispersing 1 mg of mPEG-DOPA treated magnetite in 1 ml of water (18 MΩ-cm filtered using a Millex® AP 0.22μm filter (Millipore)), DCM or Toluene. Suspensions were placed in a bath sonicator for ten minutes to disperse the nanoparticles. All three solutions were stable at room temperature for at least six months, whereas control suspensions of unmodified magnetite and magnetite stabilized by mPEG-OH or mPEG- NH2 precipitated out in less than 24 hours in each solvent. Example 14d [00178] Suspensions of mPEG-DOPA stabilized nanoparticles were also found to be stable under physiologic concentrations of salt. To determine whether mPEG-DOPA could inhibit salt-induced nanoparticle aggregation, 0.3 mg of mPEG-DOPA treated magnetite was placed in a quartz cuvette and combined with 0.7 ml of water (18 MΩ-cm filtered using a 0.25 μ filter). Aliquots of saturated NaCl solution (5 μl,10 μl, 20 μl, 50 μl, 100 μl) were sequentially added to the cuvette and allowed to stand for ten minutes before UV-VIS spectra were taken (Figure 15). The absorbance spectra of mPEG-DOPA stabilized nanoparticles suspended in solutions containing increasing NaCl concentration were nearly identical, demonstrating that mPEG-DOPA is effective at stabilizing the nanoparticles and preventing aggregation. The peak centered at 280 nm is indicative of the catechol side chain of DOPA. Example 14e [00179] The procedures and techniques illustrated in examples 14a-14d can be extended to various other metal oxide or ceramic nanoparticles, as would be understood by those skilled in the art made aware of this invention. Likewise, such applications of the present invention can further include use of a wide range of polymer-DHPD conjugates analogous to and consistent with those compositions and variations thereof described in examples 10b and l ib. As illustrated below in the preparation of semiconductor compositions, metal oxide or ceramic nanoparticles can be stabilized in situ upon formation in the presence of a polymer-DHPD conjugate of this invention. Example 15a [00180] Demonstrating stabilization of metal nanoparticles, commercial gold colloid suspension (Sigma, particle size 5 or 10 nm) was placed inside dialysis tubing (Mw cutoff of 8000 for 5nm and 15000 for 10 nm) and dialyzed in ultrapure water for 2-3 days to remove the sodium azide present in the commercial preparation. The dialyzed suspensions were then placed into small glass vials and mPEG-DOPA added (10 mg/ml). The samples were allowed to stand at room temperature for approximately 2 days, after which the samples were again dialyzed to remove excess mPEG-DOPA. Untreated 10 nm Au nanoparticles were unstable in the presence of NaCl and aggregated (Figure 16), whereas the treated Au nanoparticles remained stably suspended in the presence of aqueous NaCl (Figure 17). Example 15b [00181] Various other metal nanoparticles, including but not limited to, silver, platinum and the like can be stabilized as described in the preceding example. While stabilization was demonstrated using a representative conjugate composition of this invention, various other compositions can be prepared analogous to and consistent with the alternate embodiments described in Examples 10b and l ib. Comparable results can be obtained by in situ formation of the stabilized nanoparticles synthesized from the corresponding metal precursor in the presence of a suitable, adhesive conjugate polymer of this invention. Example 16a [00182] The data of this example demonstrates stabilization of semiconductor nanoparticles. CdS nanoparticles (quantum dots) were prepared by a standard method based on the slow mixing of dilute Cd(NO3)2 and Na2S solutions. Fresh stock solutions (2 mM) of Cd(NO3)2 and Na2S were prepared in nanopure water. The Na2S solution was injected slowly into 50 ml of Cd(NO3)2 solution using a gastight syringe at a rate of 20 μl s"1. The solution turned yellow with the addition of Na2S, and after 2 mL of Na2S was injected, a yellow precipitate appeared due to the aggregation of CdS nanoparticles. The CdS precipitate was isolated and dried for further use. Using the method described above for magnetite, the dry CdS powder was dispersed in a mPEG-DOPA solution by sonication to yield a clear yellow solution. The yellow aqueous suspension was stored in the dark for several months at room temperature without visible formation of precipitate. Control experiments performed in the absence of polymer and in the presence of mPEG-OH or mPEG-NH2 yielded yellow precipitate and a clear, colorless supernatant. mPEG-DOPA stabilized CdS nanoparticles remained stably suspended in the presence of aqueous NaCl (Figure 18). Example 16b [00183] The results of this example illustrate the in situ formation of stabilized semiconductor nanoparticles. CdS nanoparticles (quantum dots) were formed in the presence of mPEG-DOPA by slowly mixing dilute methanolic solutions of Cd(NO3)2 and Na2S. Freshly prepared stock solutions (2 mM) of Cd(NO3)2 and Na2S were prepared in methanol. 25 mg of mPEG-DOPA (PEG molecular weight=2000) was dissolved in 5 ml of 2 mM Cd(NO3)2 in methanol, then 5 ml of a 2 mM solution of Na2S was added slowly with a syringe at a rate of 20 μl s"1. The solution gradually turned yellow during the addition. No yellow precipitates were observed, and dynamic light scattering revealed particles with an average diameter of 2.5 nm. Control experiments performed in the absence of polymer or in the presence of mPEG-OH yielded yellow precipitate and a clear, colorless supernatant. Various other inorganic particulate substrates can be prepared, as would be understood by those skilled in the art, depending upon material choice and corresponding ionic substitution or exchange reaction, as carried out in the presence of an adhesive composition of the sort described herein. Example 16c [00184] The polymeric conjugate compositions of this invention can also be used to stabilize a variety of other semiconductor materials. For instance, core-shell nanoparticles can be surface stabilized in accordance herewith. Example 17 [00185] The optimization experiments of Examples 17-20 were performed with mPEG-DOPA-5K. Several parameters were examined to optimize the adsorption of mPEG-DOPA onto gold from solution, including type and pH of solvent, time of adsorption, and mPEG-DOPA solution concentration. Cell attachment and spreading did not vary widely with adsorption solvent used. The number of cells on the substrates and their total projected area was not significantly different between DCM and three different aqueous solutions. The substrates adsorbed in neutral, basic, and organic mPEG-DOPA solutions all possessed significantly enhanced anti-fouling properties when compared to the unmodified substrate (p<0.01). Although no differences were observed in cell attachment and spreading between the solutions, the contact angle data would support the use of an organic solvent in an optimal modification protocol as a means to reduce catechol oxidation. Additionally, only the surface modified in DCM demonstrated significantly fewer cells on the surface and lower total projected cellular area. Example 18 [00186] Cell attachment and spreading showed a strong dependence on solution concentration of mPEG-DOPA (Figure 12). Above 25 mg/ml mPEG-DOPA, significantly fewer cells attached and spread on the modified substrate than on the pristine gold surface (p<0.001) and the surface modified in a 10 mg/ml solution (p<0.05). Below 10 mg/ml, there were no differences in cell attachment and spreading compared to the unmodified substrate. There were no differences in cell attachment and spreading observed between surfaces modified in mPEG-DOPA solutions ranging from 25-75 mg/ml when compared to each other. Example 19 [00187] Fewer fibroblasts were observed to attach and spread with increasing duration of mPEG-DOPA adsorption, as well. Although cell attachment and spreading appeared to decrease with as little as 5 minutes of substrate modification, an adsorption time of 24 hours resulted in significantly fewer cells attaching and spreading on the PEGylated substrate than on the unmodified substrate (p<0.001) and substrates treated for shorter periods (p<0.05). Example 20 [00188] The morphology of fibroblasts cultured on both unmodified and PEG- modified surfaces was examined via electron microscopy (Hitachi 3500 SEM). Fibroblasts on unmodified Au and mPEG-OH-modified Au were generally flat and well spread, while those cultured on mPEG-DOPA modified Au were far less spread (Figures 14A-C). It should also be noted that on the mPEG-DOPA surface, a lower number of cellular processes were observed than in the others, structures which contribute to cell adhesion via integrins and focal adhesions. Figure 13 illustrates the differences in attachment and spreading of fibroblasts on bare Au, mPEG-OH-treated Au, and Au modified with mPEG-DOPA 5K, mPEG-MAPd 2K, or mPEG-MAPd 5K under optimal conditions (50 mg/ml for 24 hours). The surfaces modified with DOP A-containing conjugates have significantly less cellular adhesion and spreading than either of the other two surfaces. The mPEG-MAP 5K modification, though, accounted for a 97% reduction in total projected cellular area and a 91% reduction in density of cells on the surface, a far greater reduction than that achieved by mPEG-DOPA 2K.
[00189] The differences in cellular adhesion and spreading between surfaces mo'dified with DOPA- and MAPd-conjugated PEG in Figure 13 can likely be attributed to the physical characteristics of the associated PEG adlayer. Analysis of the SPR results indicates that MAPd-PEGs form thicker, more robust adlayers with a higher concentration of PEG per unit area than do the DOPA-anchored PEGs of equivalent molecular weight. The thicker adlayers resulting from MAPd-mediated PEGylation are more successful in inhibiting protein adsorption and, in turn, cell adhesion. Example 21
Synthesis of Boc-DOPA(TBDMS)2-OSu
[00190] N-hydroxysuccinimide (NHS) (0.110 g, 0.95 mmol) was added to a solution of Boc-DOPA(TBDMS)2 (0.500 g, 0.95 mmol) in dry dichloromethane (DCM) (8.0 mL). The solution was stirred on an ice bath, and 1,3-dicyclohexylcarbodiimide (DCC) (0.197 g, 0.95 mmol) was added under nitrogen atmosphere. The reaction was stirred for 20 minutes at 0°C and then warmed to room temperature and stirred for an additional 4 hours. The reaction mixture was filtered to remove the urea byproduct and subsequently evaporated to 1/5 of its original volume. The solution was cooled to 4°C, allowed to sit for 2 hours to precipitate the remaining urea byproduct, filtered and evaporated to give Boc-DOPA(TBDMS)2-OSu as a white foam (0.567 g, 96 % yield). Example 22 Synthesis ofBoc-DOPA2(TBDMS)4
[00191] Boc-DOPA(TBDMS)2-OSu (0.567 g, 0.91 mmol) was dissolved in dry dimethylformamide (DMF) (2.5 mL), and DOPA(TBDMS)2 (0.405 g, 0.95 mmol) was added at once under a nitrogen atmosphere. The mixture was stirred on an ice bath, and diisopropylethylamine (DIEA) (158 μL, 0,91 mmol) was added dropwise via a syringe. After 20 minutes, the reaction was warmed to room temperature, stirred for an additional 17 hours, filtered (if necessary), diluted with ethyl acetate (EtOAc) (40 mL), transferred to a separatory funnel, and washed with 5% aqueous HCl. The aqueous layer was extracted back with EtOAc. The organic layers were combined and washed with 5% aqueous HCl (3x), H2O (lx), dried with MgSO4, and evaporated to afford Boc- DOPA2(TBDMS)4 as a white foam (0.83 g, 98 % yield). Example 23 Synthesis ofBoc-DOPA2 (TBDMS) 4-OSu
[00192] The procedure of Example 21 was repeated using Boc-DOPA2(TBDMS) to obtain Boc-DOPA2(TBDMS)4-OSu. Example 24 Synthesis ofBoc-DOPA3(TBDMS)6
[00193] The procedure of Example 22 was repeated using Boc-DOPA2(TBDMS)4-
OSu to obtain Boc-DOPA3(TBDMS)6. Example 25
Synthesis ofDOPA2
[00194] Boc-DOPA2(TBDMS)4 (0.5 g, 0.54 mmol) was dissolved in saturated
HCl/EtOAc (3 mL), and the solution was stirred under nitrogen. After 5 hours, additional HCl gas was gently bubbled through the solution for 25 minutes. The reaction was allowed to sit overnight, and subsequently concentrated to lA of original volume. The resulting precipitate was collected by centrifugation, washed with cold EtOAc (3x), and dried to yield DOPA2 as a white powder (0.15 g, 74% yield). The product was further purified by preparative RP-HPLC, and characterized by ESI-MS. Example 26 Synthesis of DOP A
[00195] Boc-DOPA3(TBDMS)6 (1.06 g, 0.79 mmol) was dissolved in saturated
HCl/EtOAc (3 mL), and the solution was stirred under nitrogen. After 12 hours, additional HCl gas was gently bubbled through the solution for 30 minutes, and the reaction was allowed to continue for 40 hours. More HCl gas was bubbled through the solution for another 30 minutes, and the stirring was stopped. The resulting precipitate was collected by centrifugation, washed with cold EtOAc (3x), and dried to give DOPA3 as a white powder (0.424 g, 96% yield). The product was further purified by preparative RP-HPLC, and characterized by ESI-MS. Example 27 Synthesis ofmPEG-DOPA].3
[00196] A solution of 0.1 M borate buffer (50 mL, pH 8.5) was degassed with argon for 20 minutes, and L-DOPA (0.197 g, 1.0 mmol) was added. After the solution was stirred for 15 minutes, methoxy-terminated PEG-SPA (mPEG-SPA) 5K (0.5 g, 0.1 mmol) was added in portions, and the reaction was allowed to stir for 3 hours. The resulting clear solution was then acidified to pH of 1-2 with aqueous HCl, and extracted with DCM (3x). The combined organic layers were washed with 0.1 M HCl, dried over MgSO4 and concentrated. The remaining residue was dissolved in DCM and precipitated with ethyl ether three times to afford mPEG-DOPA as a white powder (0.420 g, 84% yield). The product was characterized by MALDI-MS and 1H NMR spectroscopy. Example 28
Surface Modification
[00197] Solid metal substrates (Al, 316L stainless steel and NiTi) were ground and polished, ultimately with 0.04 m colloidial silica (Syton, DuPont). Si wafers were evaporated with either 20 nm TiO2 or 10 nm TiO2/40 nm Au using an Edwards FL400 electron beam evaporator at <10"6 Torr and were subsequently diced in 8 mm x 8 mm pieces. All substrates were cleaned ultrasonically for 20 minutes in each of the following: 5% Contrad70 (Fisher Scientific), ultrapure H2O, acetone, and petroleum ether. Subsequently, surfaces were further cleaned by exposure to O2 plasma (Harrick Scientific) at 150 mTorr and 100 W for 5 minutes. To prevent the formation of a gold oxide (Au2O3) layer, some Au substrates were not exposed to O2 plasma. To generate a biopolymer analog surface, glass coverslips (Fisher Scientific) were cleaned as described above and immersed in a 0.01% solution of poly-L-lysine (Sigma) for 5 minutes, rinsed with ultrapure H2O, and dried under nitrogen.
[00198] In order to explore a variety of modification conditions with a minimum number of samples, a nine element Robust Design approach was employed. Substrates were modified under cloud point conditions by immersion in mPEG-DOPAι-3 solutions in 0.6 M K2SO4 buffered with 0.1 M MOPS at 50°C. Buffer pH, mpdification time, and mPEG-DOPA concentration were modified as shown in Table 1. Modified substrates were subsequently rinsed with ultrapure H2O and dried under a stream of nitrogen. Table 1
Figure imgf000066_0001
Example 28a
Ti0 Substrates
[00199] TiO2 substrates were modified under cloud point conditions by immersion in mPEG-DOPA1-3 solutions in 0.6 M K2SO4 buffered with 0.1 M N- morpholinopropanesulfonic acid (MOPS) at 50°C for 24 hours. Modified substrates were rinsed with ultrapure H2O and dried under a stream of nitrogen. Example 28b
[00200] 316L Stainless Steel (Goodfellow, Devon PA) was modified under cloud point conditions by immersion in mPEG-DOPAι-3 solutions in 0.6 M K2SO4 buffered with 0.1 M N-morpholinopropanesulfonic acid (MOPS) at 50°C for 24 hours. Modified substrates were rinsed with ultrapure H2O and dried under a stream of nitrogen. Example 28c
[00201] Al2O3 (Goodfellow, Devon PA) was modified under cloud point conditions by immersion in mPEG-DOPA1-3 solutions in 0.6 M K2SO4 buffered with 0.1 M N-morpholinopropanesulfonic acid (MOPS) at 50°C for 24 hours. Modified substrates were rinsed with ultrapure H2O and dried under a stream of nitrogen. Example 28d
[00202] SiO2 (1500A thermal oxide, University Wafer, South Boston, MA) was modified under cloud point conditions by immersion in mPEG-DOPA1-3 solutions in 0.6 M K2SO buffered with 0.1 M N-morpholinopropanesulfonic acid (MOPS) at 50°C for 24 hours. Modified substrates were rinsed with ultrapure H2O and dried under a stream of nitrogen. Example 28e
[00203] NiTi alloy (10mm x 10mm x 1mm) was obtained from Nitinol Devices &
Components (Fremont, CA) and modified under cloud point conditions by immersion in mPEG-DOPAi-3 solutions in 0.6 M K2SO4 buffered with 0.1 M N- morpholinopropanesulfonic acid (MOPS) at 50°C for 24 hours. Modified substrates were rinsed with ultrapure H2O and dried under a stream of nitrogen. Example 28f
[00204] Au (electron beam evaporated onto Si Wafer from University Wafer) was modified under cloud point conditions by immersion in mPEG-DOPA1-3 solutions in 0.6 M K2SO buffered with 0.1 M N-morpholinopropanesulfonic acid (MOPS) at 50°C for 24 hours. Modified substrates were rinsed with ultrapure H2O and dried under a stream of nitrogen. Example 28g
[00205] Au2O3 (Au samples as described in Example 28f were exposed to an oxygen plasma to form Au2O3) was modified under cloud point conditions by immersion in mPEG-DOPAι-3 solutions in 0.6 M K2SO buffered with 0.1 M N- morpholinopropanesulfonic acid (MOPS) at 50°C for 24 hours. Modified substrates were rinsed with ultrapure H2O and dried under a stream of nitrogen. Example 28h
[00206] GaAs (University Wafer, South Boston, MA) was modified under cloud point conditions by immersion in mPEG-DOPAι-3 solutions in 0.6 M K2SO4 buffered with 0.1 M N-morpholinopropanesulfonic acid (MOPS) at 50°C for 24 hours. Modified substrates were rinsed with ultrapure H2O and dried under a stream of nitrogen. Example 28i
[00207] p-L-Lys surfaces were made by immersing glass coverslips (Fisher
Scientific) in a 0.01% solution of poly-L-lysine (p-L-Lys, Sigma) for 5 minutes, rinsed with ultrapure H2O, and dried under N2. They were then modified under cloud point conditions by immersion in mPEG-DOPA1-3 solutions in 0.6 M K2SO4 buffered with 0.1 M N-morpholinopropanesulfonic acid (MOPS) at 50°C for 24 hours. Modified substrates were rinsed with ultrapure H2O and dried under a stream of nitrogen. Example 29 Cell Adhesion
[00208] 3T3 Swiss albino fibroblasts (ATCC, Manassas, VA) of passage 12-16 were cultured normally at 37°C and 5% CO2 in Dulbecco's modified Eagle's medium (DMEM) (Cellgro, Herndon, VA) supplemented with 10% fetal bovine serum (FBS) (Cellgro, Herndon, VA), 100 g/mL penicillin, and 100 U/mL steptomycin. Prior to cell adhesion assays, fibroblasts were harvested using 0.25% trypsin-EDTA, resuspended in growth medium, and counted with a hermacytometer. General Procedure for Four-Hour Assay
[00209] Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO2. Cells were seeded onto the substrates at a density of 2.9 x 103 cells/cm2 and maintained in DMEM with 10% FBS at 37°C and 5% CO2 for 4 hours. For the 4-hour cell adhesion assay, adherent cells were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently stained with 5μM l,l'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil) (Molecular Probes, Eugene, OR) in DMSO for 45 minutes at 37°C. [00210] Quantitative cell attachment data was obtained by acquiring 9-16 images
(depending on substrate size) from random locations on each substrate using a Leica epifluorescent microscope equipped with a SPOT RT digital camera (Diagnostic Instruments, Sterling Heights, MI). The resulting images were quantified in terms of total projected cellular area using thresholding in MetaMorph. (Universal Imaging Corporation1 , a subsidiary of Molecular Devices Corporation, Downington, PA). The mean and standard deviation of the measurements are reported. Example 29a TiO2 substrate (4-hour assay)
[00211] Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO2. Cells were seeded onto the substrates at a density of 2.9 x 103 cells/cm2 and maintained in DMEM with 10% FBS at 37°C and 5% CO2 for 4 hours. For the 4-hour cell adhesion assay, adherent cells were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently stained with 5μM l,r-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil) (Molecular Probes, Eugene, OR) in DMSO for 45 minutes at 37°C. [00212] Quantitative cell attachment data was obtained by acquiring 9-16 images
(depending on substrate size) from random locations on each substrate using a Leica epifluorescent microscope equipped with a SPOT RT digital camera (Diagnostic Instruments, Sterling Heights, MI). The resulting images were quantified in terms of total projected cellular area using thresholding in MetaMorph. (Universal Imaging Corporation™, a subsidiary of Molecular Devices Corporation, Downington, PA). The mean and standard deviation of the measurements are reported. Example 29b
Ti0 substrate (long-term studies)
[00213] For long-term studies on TiO2, substrates were reseeded twice per week at the same density as for the 4-hour assay. At periodic intervals, non-adherent cells were removed by aspirating the medium in each well. Example 29c 316L Stainless Steel substrate (4-hour assay)
[00214] Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO2. Cells were seeded onto the substrates at a density of 2.9 x 103 cells/cm2 and maintained in DMEM with 10% FBS at 37°C and 5% CO2 for 4 hours. For the 4-hour cell adhesion assay, adherent cells were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently stained with 5μM l,l'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil) (Molecular Probes, Eugene, OR) in DMSO for 45 minutes at 37°C. [00215] Quantitative cell attachment data was obtained by acquiring 9-16 images
(depending on substrate size) from random locations on each substrate using a Leica epifluorescent microscope equipped with a SPOT RT digital camera (Diagnostic Instruments, Sterling Heights, MI). The resulting images were quantified in terms of total projected cellular area using thresholding in MetaMorph. (Universal Imaging Corporation™, a subsidiary of Molecular Devices Corporation, Downington, PA). The mean and standard deviation of the measurements are reported. Example 29d Al2O3 substrate (4-hour assay)
[00216] Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO2. Cells were seeded onto the substrates at a density of 2.9 x 103 cells/cm2 and maintained in DMEM with 10%) FBS at 37°C and 5% CO2 for 4 hours. For the 4-hour cell adhesion assay, adherent cells were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently stained with 5μM l,l '-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil) (Molecular Probes, Eugene, OR) in DMSO for 45 minutes at 37°C. [00217] Quantitative cell attachment data was obtained by acquiring 9-16 images
(depending on substrate size) from random locations on each substrate using a Leica epifluorescent microscope equipped with a SPOT RT digital camera (Diagnostic Instruments, Sterling Heights, MI). The resulting images were quantified in terms of total projected cellular area using thresholding in MetaMorph. (Universal Imaging Corporation™, a subsidiary of Molecular Devices Corporation, Downington, PA). The mean and standard deviation of the measurements are reported. Example 29e SiO2 substrate (4-hour assay)
[00218] Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO2. Cells were seeded onto the substrates at a density of 2.9 x 103 cells/cm2 and maintained in DMEM with 10% FBS at 37°C and 5% CO2 for 4 hours. For the 4-hour cell adhesion assay, adherent cells were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently stained with 5μM l,r-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil) (Molecular Probes, Eugene, OR) in DMSO for 45 minutes at 37°C. [00219] Quantitative cell attachment data was obtained by acquiring 9-16 images
(depending on substrate size) from random locations on each substrate using a Leica epifluorescent microscope equipped with a SPOT RT digital camera (Diagnostic Instruments, Sterling Heights, MI). The resulting images were quantified in terms of total projected cellular area using thresholding in MetaMorph. (Universal Imaging Corporation™, a subsidiary of Molecular Devices Corporation, Downington, PA). The mean and standard deviation of the measurements are reported. Example 29f NiTi substrate (4-hour assay)
[00220] Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO2. Cells were seeded onto the substrates at a density of 2.9 x 103 cells/cm2 and maintained in DMEM with 10% FBS at 37°C and 5% CO2 for 4 hours. For the 4-hour cell adhesion assay, adherent cells were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently stained with 5μM l,l '-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil) (Molecular Probes, Eugene, OR) in DMSO for 45 minutes at 37°C. [00221] Quantitative cell attachment data was obtained by acquiring 9-16 images
(depending on substrate size) from random locations on each substrate using a Leica epifluorescent microscope equipped with a SPOT RT digital camera (Diagnostic Instruments, Sterling Heights, MI). The resulting images were quantified in terms of total projected cellular area using thresholding in MetaMorph. (Universal Imaging Corporation™, a subsidiary of Molecular Devices Corporation, Downington, PA). The mean and standard deviation of the measurements are reported. Example 29g Au substrate (4-hour assay)
[00222] Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO2. Cells were seeded onto the substrates at a density of 2.9 x 103 cells/cm2 and maintained in DMEM with 10% FBS at 37°C and 5% CO2 for 4 hours. For the 4-hour cell adhesion assay, adherent cells were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently stained with 5μM l,l'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil) (Molecular Probes, Eugene, OR) in DMSO for 45 minutes at 37°C. [00223] Quantitative cell attachment data was obtained by acquiring 9-16 images
(depending on substrate size) from random locations on each substrate using a Leica epifluorescent microscope equipped with a SPOT RT digital camera (Diagnostic Instruments, Sterling Heights, MI). The resulting images were quantified in terms of total projected cellular area using thresholding in MetaMorph. (Universal Imaging Corporation™, a subsidiary of Mojecular Devices Corporation, Downington, PA). The mean and standard deviation of the measurements are reported. Example 29h Au2O3 substrate (4-hour assay)
[00224] Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO2. Cells were seeded onto the substrates at a density of 2.9 x 103 cells/cm2 and maintained in DMEM with 10% FBS at 37°C and 5% CO2 for 4 hours. For the 4-hour cell adhesion assay, adherent cells were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently stained with 5μM l,l '-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil) (Molecular Probes, Eugene, OR) in DMSO for 45 minutes at 37°C. [00225] Quantitative cell attachment data was obtained by acquiring 9-16 images
(depending on substrate size) from random locations on each substrate using a Leica epifluorescent microscope equipped with a SPOT RT digital camera (Diagnostic Instruments, Sterling Heights, MI). The resulting images were quantified in terms of total projected cellular area using thresholding in MetaMorph. (Universal Imaging Corporation™, a subsidiary of Molecular Devices Corporation, Downington, PA). The mean and standard deviation of the measurements are reported. Example 29i GaAs substrate (4-hour assay)
[00226] Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO2. Cells were seeded onto the substrates at a density of 2.9 x 103 cells/cm2 and maintained in DMEM with 10%) FBS at 37°C and 5% CO2 for 4 hours. For the 4-hour cell adhesion assay, adherent cells were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently stained with 5μM l,l'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil) (Molecular Probes, Eugene, OR) in DMSO for 45 minutes at 37°C. [00227] Quantitative cell attachment data was obtained by acquiring 9-16 images
(depending on substrate size) from random locations on each substrate using a Leica epifluorescent microscope equipped with a SPOT RT digital camera (Diagnostic Instruments, Sterling Heights, MI). The resulting images were quantified in terms of total projected cellular area using thresholding in MetaMorph. (Universal Imaging Corporation1 , a subsidiary of Molecular Devices Corporation, Downington, PA). The mean and standard deviation of the measurements are reported. Example 29i p-L-Lys substrate (4-hour assay)
[00228] Test substrates were prepared in 12-well tissue culture polystyrene plates with 1.0 mL DMEM with FBS for 30 minutes at 37°C and 5% CO2. Cells were seeded onto the substrates at a density of 2.9 x 103 cells/cm2 and maintained in DMEM with 10%) FBS at 37°C and 5% CO2 for 4 hours. For the 4-hour cell adhesion assay, adherent cells were fixed in 3.7% paraformaldehyde for 5 minutes and subsequently stained with 5μM l, -dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil) (Molecular Probes, Eugene, OR) in DMSO for 45 minutes at 37°C. [00229] Quantitative cell attachment data was obtained by acquiring 9-16 images
(depending on substrate size) from random locations on each substrate using a Leica epifluorescent microscope equipped with a SPOT RT digital camera (Diagnostic Instruments, Sterling Heights, MI). The resulting images were quantified in terms of total projected cellular area using thresholding in MetaMorph. (Universal Imaging Corporation™, a subsidiary of Molecular Devices Corporation, Downington, PA). The mean and standard deviation of the measurements are reported. Example 30 Twenty-four-hour modification of substrates and four-hour assay
[00230] Each surface was modified for 24 hours in a solution of 1.0 mg/mL mPEG-DOPA3 (or mPEG-OH as a control) at 50°C at the pH values shown in Figure 24. A four-hour cell adhesion and spreading assay was conducted as described above in Example 9. Results are shown in Figure 24. Cell adhesion resistance was conferred to all substrates treated with mPEG-DOPA3. Cell adhesion and spreading on substrates treated with mPEG-OH did not differ from the unmodified surfaces (data not shown). Example 31 Surfaces and Surface Preparation.
[00231] Silicon wafers (WaferNet GmbH, Germany) were coated with TiO2 (20 nm) by physical vapor deposition using reactive magnetron sputtering (PSI, Villigen, Switzerland). Metal oxide coated wafers were subsequently diced into 1 cm x 1 cm pieces for ex-situ ellipsometry measurements. Optical waveguide chips for OWLS measurements were purchased from Microvacuum Ltd. (Budapest, Hungary) and consisted of a AF45 glass substrate (8 x 12 x 0.5 mm) and a 200 nm-thick Sio.25Tio.75O2 waveguiding surface layer. An 8 nm TiO2 layer was deposited on top of the waveguiding layer under the same conditions described above for the silicon wafers. Prior to polymer modification, TiO2-coated silicon wafers and waveguide chips were sonicated in 2-propanol for 10 minutes, rinsed with ultrapure water, and dried under a stream of nitrogen, followed by a 3 minute exposure to O2 plasma (Harrick Scientific, Ossining, USA) to remove all organic components from the surface. After OWLS measurements, waveguides were regenerated for reuse by sonication (10 minute) in cleaning solution (300 mM HCl, 1% detergent; Roche Diagnostics, Switzerland) and subsequent rinsing with ultrapure water to remove adsorbates. Surface Modification..
[00232] Surfaces were modified by mPEG-DOPA1-3; produced according to
Example 27, for 24 hours at a polymer concentration of 1.0 mg/ml using cloud point buffer (CP buffer: 0.6 M K2SO4 buffered to pH = 6.0 with 0.1 M MOPS) at temperatures ranging from 25°C to 50°C. After modification, substrates were rinsed with water, dried in a stream of N2, and immediately analyzed as described below. X-ray Photoelectron Spectroscopy (XPS) Measurements.
[00233] Survey and high resolution spectra were collected on a SAGE 100
(SPECS, Berlin, Germany) using a standard (non-monochromatized) AlKα X-ray source operating at 325 W (13 kV, 25 mA) and a take-off angle of 0°, defined as the angle between the photoelectron detector and the surface normal. Pass energies of 50 eV and 14 eV were used for survey and high-resolution spectra, respectively. The pressure of the analysis chamber remained below 2 x 10"8 Pa during data acquisition. All XPS spectra were referenced to the aliphatic hydrocarbon component of the Cis signal at 284.7 eV. Curve fitting was performed with CasaXPS software using Shirley background subtraction and the sum of a 90% Gaussian and 10% Lorentzian function. Measured intensities (peak areas) were converted to normalized intensities by atomic sensitivity factors, from which atomic compositions of surfaces were calculated. Average values obtained from three substrate replicates is reported in Tables 7-8. Standard deviations were typically <10% of the mean and are omitted for clarity. Table 7 Quantitative Analysis of XPS Data for mPEG-DOPA Modified TiO2 Surfaces atomic concentration (atom %) Ti O C OlSA Ol SB Olsc CI SA ClSB Clsc surface TiO2 TiOH C-O, H2O C-C,C-H C-O NHC(=O) Clean TiO2 24.3 50.9 14.1 4.1 4.5 1.2 0.9 mPEG- 17.9 36.8 5.2 13.1 2.8 23.1 1.2 DOPAi mPEG- 11.1 22.5 3.2 20.2 3.7 37.3 2.1 DOPA2 mPEG- 7.4 15.9 0.5 25.3 4.0 43.7 3.2 DOPA3
Table 8 Atomic Ratios Calculated from XPS Data for mPEG-DOPA Modified TiO2 Surfaces atomic ratioa surface C/Ti CVrX O CVC Cc/C Cc/C A A B A clean TiO2 0.27 2.10 0.28 0.33 0.42 0.20 mPFG- ™Qp^ 1.52 2.06 0.14 8.42 0.052 0.42 mPFG- τ DX™Ur AI 3.89 2.03 0.14 10.07 0.055 0.56 DO«P?A~ 3 6-87 2-!4 0.03 11.000.074 0.80 a Contributions A, B, and C are defined in Table 7.
Spectroscopic Ellipsometry.
[00234] For ELM measurements, TiO2-sputtered Si substrates were modified ex- situ as described above, with the temperature of the modification solution varied from 25 °C to 50 °C. After modification, substrates were rinsed with H2O, incubated at room temperature in 10 mM HEPES buffer (pH = 7.4) for 48 hours, rinsed again with H2O and dried with N2. To examine protein resistance, modified and unmodified substrates were exposed to pure human serum for 15 minutes, rinsed with water, and dried in a stream of N2. ELM measurements were made on a M-2000D spectroscopic ellipsometer (J. A. Woollam Co., Inc., Lincoln, USA) at 65°, 70°, and 75° using wavelengths from 193 - 1000 nm prior to and immediately after modification, after HEPES incubation, and after serum exposure. ELM spectra were fit with multilayer models in the WVASE32 analysis software using the optical properties of a generalized Cauchy polymer layer (An = 1.45, B„ = 0.01. Cn = 0), to obtain the "dry" thicknesses of adsorbed PEG and serum adlayers. (The "dry" or dehydrated thickness is that measured under ambient conditions after drying with N2.) The average thickness measured from three replicates is reported in Tables 9-10. Table 9 Effect of Adsorption Temperature on Thickness of PEG Adlayers on TiO2 a Thickness (A) Temperature (°C) 25 10.6 ± 1.8 28 13.9 ± 0.3 31 16.4 ± 1.1 34 19.4 ± 1.9 37 19.6 ± 3.5 40 22.5 ± 2.0 45 24.4 ± 2.0 50 33.8 ± 4.6 a TiO2 surfaces were exposed to mPEG-DOPA3 (lmg/ml) for 2h after which each surface was rinsed for 48h in HEPES.
Table 10 Apparent Thickness (A) of Organic Adlayers on TiO2 as Measured by Spectroscopic Ellipsometry mPEG-DOPA3 adsorption time (min) Treatment" 0 1 30 60 240 1080
Before serum exposure - 5.1 24.2 27.6 31.8 35.0
After serum exposure 60.2 23.4 24.7 <28.1* <32.3* <35.5ό α TiO2 surfaces were exposed to mPEG-DOPA3 (lmg/ml) for 0-1080min, rinsed with water, incubated for 48h in HEPES and then exposed to serum for 15 min. The net increase in adlayer thickness after serum exposure was less than 0.5 A, the approximate resolution of the ELM technique.
Optical Waveguide Lightmode Spectroscopy (OWLS).
[00235] TiO2-coated waveguides were cleaned in 2-propanol and O2 plasma as described above. Clean waveguides were mounted in the measurement head of an OWLS110 (Microvacuum Ltd.) and stabilized for at least 48 hours at room temperature in cloud point buffer (CP buffer: 0.6 M K2SO4 buffered to pH = 6.0 with 0.1 M MOPS). The stabilization period allowed for the exchange of ions at the TiO2 surface to come to equilibrium and obtain a stable baseline. To monitor polymer adsorption, mPEG-DOPA in CP buffer was injected in stop-flow mode, followed by CP buffer to remove unbound PEG, after which the signal was allowed to stabilize. The incoupling angles, ( TM and ( TE, were recorded and converted to refractive indices (NiM, Nre) by the manufacturer- supplied software. Real-time changes in the effective refractive index of the sensor were converted to adsorbed mass using de Feijter's formula. Need details of reference for incorporation[de Feijter, 1978 #14] The refractive index increment, άnlάc, for each mPEG-DOPA polymer was calculated by linear interpolation between 0.13 cm3/g for pure PEG and 0.18 cm3/g for pure poly(amino acid). For protein adsorption experiments, the temperature of the measurement head was equilibrated at 37 °C until the signal stabilized, after which serum was injected for 15 minutes followed by injection of buffer. Substantial differences in adsorbed mass were not observed with increases in serum exposure time. Example 32 Synthesis ofN-methacryloyl 3, 4-dihydroxyl-L-phenylalanine
[00236] 1.15 g (5.69 mmol) of Na2B4O7 was dissolved in 30 ml of water. The solution was degassed with Ar for 30 minutes., after which 0.592 g (3.0 mmol) of L- DOPA was added and stirred for 15 minutes. 0.317 g (3.0 mmol) of Na2CO3 was then added, the solution was cooled to 0°C, and 0.3 ml (3.0 mmol) of methacryloyl chloride was slowly added with stirring. The pH of the solution was maintained above 9 with Na CO3 during the reaction. After stirring for 1 hour at room temperature, the solution was acidified to pH of 2 with concentrated HCl. The mixture was extracted with ethyl acetate three times. After washing with 0.1 N HCl and drying over anhydrous MgSO4, the solvent was removed in vacuo to yield crude light brown solid. The product was further purified by elution from a silica gel column with dichloromethane (DCM) and methanol (95:5). After evaporating the solvent, a white, sticky solid was obtained with a product yield of 35%. 1H NMR (500 MHz, acetone-^): y 7.1 d (1H,-NH-); 6.6-6.8 (3Η, C6H3(OH)2-); 5.68 s (IH, CHH=); 5.632 s (unknown peak); 5.33 s (IH, CHH=); 4.67 m (1Η, -CH-); 2.93-3.1 m (2Η, CH2-); 1.877 s (3 Η, -CH3). Example 33 Synthesis of 3 ,4-bis(t-butyldimethylsilyloxy)-L-phenylalanine
[00237] 3.60 g (24.0 mmol) of TBDMS-C1 was dissolved in 18 ml of anhydrous acetonitrile. 1.60 g (8.0 mmol) of L-DOPA was added to the solution, the suspension was stirred and cooled to 0°C, and 3.6 ml of DBU (24.0 mmol) was added. The reaction mixture was then stirred for 24 hours at room temperature. Addition of cold acetonitrile to the reaction solution resulted in a colorless precipitate. The precipitate was filtered and washed with cold acetonitrile several times followed by drying in vacuum. White powder was obtained with a yield of 78%. 1H NMR (500 MHz, methanol- ); y 6.7-6.9 (e H, C6H3(O-Si-)2-); 3.72 (m, 1 H, -CH-); 2.82-3.2 (m, 2 Η, -CH2-); 1.0 (d, 18 Η, - C(CH3)); 0.2 (d, 12 Η, Si-CH3). Example 34 Synthesis of3,4-bis(t-butyldimethylsilyloxy)-N-t-butyloxycarbonyl-L-phenylalanine [00238] 1.60 g (3.77 mmol) of 3,4-bis(t-butyldimethylsilyloxy)-L-phenylalanine was added to 10 ml of water containing 0.34 g (4.05 mmol) of NaΗCO3. 0.96 g (4.30 mmol) of di-t-butyl dicarbonate in 10 ml of tetrahydrofuran was added and the reaction mixture was stirred for 24 hours at room temperature. After evaporation of tetrahydrofuran, 10 ml of water was added to the residue. The solution was acidified with dilute HCl to pH of 5 and extracted three times with ethyl acetate. After drying over anhydrous MgSO4, the solvent was removed in vacuo. The crude product was purified by column chromatography (silica gel; eluent; 10% methanol in DCM). A white solid was obtained with a yield of 70% after evaporating the eluting solvent. 1H NMR (500 MHz, methanol- d); y 6.68-6.81(3 H, C6H3(O-Si-)2-); 4.28 (m, 1 H, -CH-); 2.78-3.08 (m, 2 Η, -CH3-); 1.4 (s, 9 Η, -O-C(CH3)3); 1.0 (d, 18 Η, -Si-C(CH3)3); 0.2 (d, 12 Η, Si- (CH3)2). Example 35 Synthesis of3,4-Bis(t-butyldimethylsilyloxy)-N-t-butyloxycarbonyl-L-phenylalanine Pentafluorophenyl Ester
[00239] 1 g (1.90 mmol) of 3,4-bis(t-butyldimethylsilyloxy)-N-t- butyloxycarbonyl-L-phenylalanine and 0.351 g (1.90 mmol) of pentafluorophernol were dissolved in a solvent mixture of 24 ml of dioxance and 1 ml of DMF, and 0.432 g (2.10 mmol) of DCC was added at 0°C. The solution was stirred for 1 hour at 0°C and for 1 hour at room temperature, after which the solution was filtered to remove dicyclohexylurea and evaporated in vacuo. The product 4 was purified by column chromatography (silica gel; eluent; hexane/ethyl acetate=11.2). After removing the eluent, pure white, sticky solid was obtained with a yield of 55%. 1H NMR (500 MHz, CDC13); y 6.65-6.81(3 H, C6H3(O-Si-)2-); 4.85 (m, 1 H, -CH-); 3.05-3.2 (m, 2 Η, -CH3-); 1.41 (s, 9 Η, -O-C(CH3)3); 1.0 (d, 18 Η, -Si-C(CH3)3); 0.2 (d, 12 Η, Si-(CH3)2). Example 36
Synthesis ofN-(13 '-Amino-4 ', 7 ',10 '-trioxatridecanyl)-t-butyloxycarbonyl-3 ' , 4 '-bis(t- butyldimethylsilyloxy)-L-phenylalanamide
[00240] 0.869 g (1.26 mmol) of 3,4-Bis(t-butyldimethylsilyloxy)-N-t- butyloxycarbonyl-L-phenylalanine pentafluorophenyl ester in 10 ml of DCM was added dropwise to a mixture of 2.07 ml (9.44 mmol) of 4,7,10-trioxa-l,13-tridecanediamine and 1.32 ml (9.44 mmol) of Et3N in 1 ml DMF over 30 minutes at 0°C. The solution was stirred at room temperature for another 2 hours, and then the solvent was removed under vacuum. The crude product was loaded onto silica gel and eluted with DCM, 5% methanol in DCM, 10% methanol in DCM, and 15% methanol in DCM. The solvent was removed under vacuum to yield 5 as a white solid. The yield was 63%. 1H NMR (500 MHz, acetone-d6); y 7.38 (m, 1 H, -CONH-); 6.60-6.80 (3 Η, C6Η3(O-Si-)2-); 5.26 (m, 1 H, -CONH-); 4.30 (m, 1 Η, -CH-); 3.4-3.8 (m, 12 Η, -CH2O-; 3.03-3.4 (m, 4 Η, - CH2-NΗ-, -CH2-NH2); 2.78-3.02(m, 2 H, -CH2-); 2.0 (m, 2 Η, -CH2-); 1.7 (m, 2 Η, -CH2- ); 1.39 (s, 9 Η, -O- C(CH3)3); 1.0 (d, 18 Η, Si-C(CH3)3); 0.2 (d, 12 Η, Si-C(CH3)2).
Example 37
[00241] Synthesis ofN-(13-(N'-t-Butyloxycarbonyl- L-Amino-3 ', 4 '-bis(t- butyldimethylsilyloxy)-4, 7, 10-trioxatridecanyl)-methacrylamide [00242] 0.57 g (0.79 mmol) of N-(13'-Amino-4',7',10'-trioxatridecanyl)-t- butyloxycarbonyl-3' ,4'-bis(t-butyldimethylsilyloxy)-L-phenylalanamide and 0.166 ml (1.18 mmol) of Et3N were dissolved in 5 ml of anliydrous chloroform, to which 0.176 ml (1.18 mmol) of methacrylic anhydride was added. The solution was stirred at room temperature for 3 hours, then solvent was removed in vacuo. Pure 6 was obtained by column chromatography (silica gel; eluent: ethyl acetate) as a white, sticky solid with a yield of 61%. 1H NMR (500 MHz, CDC13); y 6.60-6.80 (3 H, C6H3(O-Si-)2-); 6.40 (m, 1 H, -CONH-); 5.71 s (1Η, CHΗ=); 5.30 s (IH, CHH=); 5.096 (m, 1 Η, -CONH-); 4.21 (m, 1 Η, -C H-); 3.2-3.65 (m, 16 Η, -CH2O, -CH2-NΗ~CH2-NΗ2); 2.80-2.99 (m, 2 H, -CH2-); 1.96 s (3 Η, -CH3); 1.81 (m, 2 Η, -CH2-); 1.68 (m, 2 Η, - CH2-); 1.40 (s, 9 Η, -O- C(CH3)3); 1.0 (d, 18 Η, Si-C(CH3) 3); 0.2 (d, 12 Η, Si-C(CH3)2). Example 38 [00243] Synthesis ofN-(13-(N'-t-Boc-L-3 ', 4 'dihydroxylphenylalaninamido)-
4, 7, 10-trioxatridecanyl)-methacrylamide
[00244] To a 10 ml round bottom flash were added 0.344 g (0.433 mmol) of N-
( 13-(N' -t-Butyloxycarbonyl- L-Amino-3 ' ,4 ' -bis(t-butyldimethylsilyloxy)-4,7, 10- trioxatridecanyl)-methacrylamide , 3 mL of TΗF, and 0.137 g (0.433 mmol) of TBAF. The solution was stirred at room temperature for 5 minutes, then 3 ml of 0.1 N ΗCI was added. The solution was extracted three times with DCM, after which the solvent was evaporated in vacuum. 7 was obtained as a white solid by column chromatography (silica gel; eluent: 7% methanol in DCM) with a yield of 63%. 1H NMR (500 MHz, acetone-de); y 7.90 (m, 1 H, -CONH-); 7.23-7.40 (d 2 Η, C6ΗH2(OΗ)2-); 6.56-6.76 (3 H, C6HH2(OH)2-; 5.930 (m, 1 H, -CONH-); 5.71 s (1Η, CHΗ=); 5.30 ^ (IH, CHH=); 4.20 (m, 1 Η, -CH-); 3.1-3.60 (m, 16 Η, -CH2O, -CH2-NΗ-,-CH2-NΗ2); 2.70-2.95 (m, 2 H, -CH2-); 1.96 5 (3 Η, -CH3); 1.78 (m, 2 Η, -CH2-); 1.65 (m, 2 Η, - CH2-); 1.39 (s, 9 Η, -O- C(CH3)3). Example 39 Synthesis of PEG-Diacrylate (PEG-DA)
[00245] 40 g (5 mmol) of PEG was dried by azeotropic evaporation in benzene and then dissolved in 150 mL of DCM. 4.18 mL (30 mmol) of Et3N and 3.6 mL (40 mmol) of acryloyl chloride was added to the polymer solution. The mixture was refluxed with stirring for 5 hours and allowed to cool at room temperature overnight. Ether was added to the mixture to form a faint yellow precipitate. The crude product was then dissolved in saturated NaCl solution, which was heated to 60°C to form two layers. DCM was added to the top layer and MgSO4 was added to remove moisture. After filtration of MgSO , the volume of the solvent was reduced in vacuum and the sample was precipitated in ether. The final product was dried in vacuum and stored at -15°C. The yield was 75%. 1H NMR (500 MHz, D2O): δ 6.47 (d, 1 H, CHH=C-); 6.23 (m, 1 H, C=CH-C(=O)-O-); 6.02 (d, 1 Η, CΗH=C-); 4.35 (m, 2'Η, -CH2-O-C(=O)-C=C); 3.23- 3.86 (PEG CΗ2) Example 40 Photopolymerization of PEG-DA
[00246] Precursor solutions of PEG-DA, 1, 7, and photoinitiator were prepared and mixed immediately before photopolymerization. Stock solutions of PEG-DA (200 mg/mL) and! (40 mg/mL) were dissolved in N2-purged phosphate buffered saline (PBS, pH 7.4), where 7 (60 mg/mL) was dissolved in 50:50 PBS/95% ethanol previously purged with N2. To prepare the final polymerization mixture, solutions of 1 or 7 were combined with PEG-DA to achieve a final concentration of PEG-DA and DHPD derivatives of 150 mg/mL. 100 μL of mixture was then added to a disc-shaped mold (100 μL, diameter = 9 mm, depth = 2.3 mm, Secure Seal® SA8R-2.0, Grace Bio Lab, Inc., OR) and irradiated for up to 20 minutes either with an UV lamp (Black Ray® Lamp, 365 nm, Model UVL-56, UVP, CA) or a blue light lamp (VIP®, 400-500 mn, BISCO Inc., IL). For UV initiated photocuring, DMPA (600 mg/mL in VP) was added to the polymeric solution to make a final concentration of 34 mM. Visible light-induced curing was performed using either CQ (100 mg/mL in VP, final concentration = 150 mM) with DMAB (30 mg/mL in VP, final concentration = 151 mM), or FNa2 (188 mg/mL in PBS, final concentration = 2 mM) with AA (100 mg/mL in PBS, final concentration = 17 mM) as the photoinitiator. The final VP concentration was adjusted to be between 135 and 300 mM.
[00247] After irradiation, the gels were blotted with filter paper to remove the liquid surface layer and weighed. Percent gel conversion was then determined by dividing the weight of the gel by the weight of 100 μL of the precursor solution. Example 41
Determining DOPA incorporation
[00248] The amount of DOPA incorporated into the photopolymerized gel was determined using a modification of the colorimetric DOPA assay developed by Waite and Benedict. Photocross-linked gels were stirred in 3 mL of 0.5 N HCI to extract DOPA monomers that were not incorporated into the gel network. 0.9 mL of the nitrite reagent (1.45 M sodium nitrite and 0.41 M sodium molybdate dihydrate) and 1.2 mL of IM NaOH were added to 0.9 mL of the extraction solution, and the absorbance (500 nm) of the mixture were recorded using a Hitachi U-2010 UV-Vis spectrophotometer with 2 to 4 minutes of NaOH addition. Standard curves were constructed using known 1 to 7 concentrations. Example 42 Mechanical Test
[00249] Hydrogels were formed in the shape of a hemisphere by loading 25 μL of the polymer mixture onto a glass slide treated with lH,lH,2H,2H-perfluorooctyltrichlorosilane. Gels were irradiated for 10 minutes, dialyzed in 0.15 M HCl for at least 24 hours to extract unincorporated DOPA monomers, and then equilibrated in PBS for greater than 15 minutes prior to testing. To determine the gel modulus, hemispherical gel caps were attached to one end of a steel cylinder (diameter = 6 mm, length = 30 mm) using superglue. The other end of the cylinder was attached to a piezoelectric stepping motor (IW-701-00, Burleigh Instruments, NY) aligned in series with a 50 g load transducer (FTD-G-50, Schaevitz Sensors, VA) with a resolution of approximately 0.1 mN. A fiber optic displacement sensor (RC100-GM2OV, Philtec, Inc., MD) measured the axial movement of the steel rod. A TiO2-coated Si wafer was positioned below the hydrogel, and the Tio2 surface was flooded with PBS in order to maintain the hydration of the gel. The indenter was advanced at 5 μm/s until a maximum compressive load of 4 mN was measured. [00250] Elastic moduli were calculated by assuming Hertzian mechanics for the specific case of non-adhesive contact between an incompressible elastic hemisphere and a rigid plane, in which case the Hertzian relationship between load (Eh) and displacement (δn) becomes: 16R//2 E
Figure imgf000084_0001
(1) 9 where R and E are the radius of curvature and the elastic modulus of the hemispherical gel, respectively. The radius of curvature of the gels was determined from height and width measurements obtained from a photograph of the gel. Example 43 Chemical oxidation of PEG-DOPA into a hydrogel
[00251] 4-arm-PEG-amine (PEG-(NH2)4, Mw = 10,000) was purchased from
SunBio, Inc. (Walnut Creek, CAv) while linear PEG-bis-amine (PEG-(NH2)2, Mw =
3,400) and methoxy-PEG-amine (mPEG-NH2, Mw = 5,000) were purchased from Shearwater Polymers, Inc. (Huntsville, AL). Sephadex® LH-20 was obtained from Fluka (Milwaukee, WI). N-Boc-L-DOPA dicyclohexylammonium salt, sodium periodate (ΝaIO4), mushroom tyrosinase (MT, EC 1.14.18.1), and horseradish peroxidase (HRP, EC 1.11.1.17) were acquired from Sigma Chemical Company (St. Louis, MO). Triethylamine (Et3N), hydrogen peroxide (30 wt%, H2O2), sodium molybdate dihydrate, and sodium nitrite were purchased from Aldrich Chemical Company (Milwaukee, WI). L-Dopa was purchased from Lancaster (Windham, NH). 1-Hydroxybenzotriazole (HOBt) was obtained from Novabiochem Corp. (La Jo 11a, CA) and O-(Benzotriazol-l- yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HBTU) was acquired from Advanced ChemTech (Louisville, KY). Synthesis ofDOPA-ModifiedPEG [00252] Linear and branched DOPA-modified PEG's containing up to four DOPA endgroups were synthesized using standard carbodiimide coupling chemistry as described below. The structure of the four DOPA-modified PEG's are shown in Figure 1.
[00253] Synthesis of PEG-(N-Boc-DOPA)4, I. PEG-(NH2)4 (6.0g, 0.60 mmoles) was reacted with N-Boc-L-DOPA dicyclohexylammonium salt (4.8 mmoles), HOBt (8.0 mmoles), and Et3Ν (8.0 mmoles) in 60 mL of a 50:50 mixture of dichloromethane (DCM) and dimethylformamide (DMF). HBTU (4.8 mmoles) in 30 mL of DCM was then added and the coupling reaction was carried out under argon at room temperature for one hour. The solution was successively washed with saturated sodium chloride solution, 5% NaHCO3, diluted HCl solution, and distilled water. The crude product was concentrated under reduced pressure and purified by column chromatography on Sephadex® LH-20 with methanol as the mobile phase. The product was further purified by precipitation in cold methanol three times, dried in vacuum at room temperature, and stored under nitrogen at -20°C. 1H NMR (500 MHz, CDC13/TMS): δ 6.81-6.77 (m, 2H, C6HH2(OH)2-), 6.6 (d, IH, C6H2H(OH)2-), 6.05 (br, s, IH), 5.33 (br, s, IH), 4.22 (br, s, IH, C6H3(OH)2-CH2-CH(N-)-C(O)N-), 3.73-3.41 (m, PEO), 3.06 (m, 2Η, PEO-CH2-N-
C(O)-), 2.73 (t, 2Η, C6H3(OH)2-CH2-CH-), 1.44 (s, 9 H, (CH3)3C-). GPC-MALLS: Mw
Figure imgf000085_0001
[00254] Synthesis of PEG- (DOPA) 4, II. 3.0 g of I (0.25 mmoles) was dissolved in
15 mL of DCM at room temperature. 15 mL of TFA was added to the mixture to react for 30 minutes under argon. After evaporating the solvent in a rotary evaporator, the product was precipitated with cold methanol three times, dried in vacuum at room temperature, and stored under nitrogen at -20°C. 1H NMR (500 MHz, D2O): δ 6.79 (d, IH, C6H2H(OH)2-), 6.66 (s, IH, C6H2H(OH)2-), 6.59 (d, IH, C6H2H(OH)2-), 4.00 (t, IH, C6H3(OH)2-CH2-CH(N-)-C(O)N-), 3.70-3.34 (M, PEO), 3.24 (m, 2Η, PEG-CH2-N-C(O)-
), 3.01-2.88 (m, 2Η, C6H3(OH)2-CH2-CH(N-)-C(O)N-). GPC-MALLS: Mw = 11,400,
Figure imgf000085_0002
[00255] Synthesis ofPEG-(N-Boc-DOPA)2, III. PEG-(NH2)2 (5.0 g, 1.5 mmoles),
N-Boc-L-DOPA dicyclohexylammonium salt (5.9 mmoles), HOBt (9.8 mmoles), and Et Ν (9.8 mmoles) were dissolved in 50 mL of a 50:50 mixture of DCM and DMF. HBTU (5.9 mmoles) in 25 mL of DCM was then added, and the reaction was carried out under argon at room temperature for 30 minutes. Recovery and purification of the product was performed as described above for I. 1H NMR (500 MHz, CDC13/TMS): δ 6.81-6.77 (m, 2H, C6HH2(OH)2-), 6.59 (d, IH, C6H2H(OH)2-), 6.05 (br, s, IH), 5.33 (br, s, IH), 4.22 (br, s, IH, C6H3(OH)2-CH2-CH(N-)-C(O)N-), 3.73-3.42 (M, PEO), 3.06 (m, 2Η, PEO-CH2-N-C(O)-), 2.74 (t, 2Η, C6H3(OH)2-CH2-CH(N-)-C(O)N-), 1.44 (s, 9 H,
(CH3)3CO-). GPC-MALLS: Mw = 4,600, Mw /Mn = 1.02.
[00256] Synthesis of methoxy-PEG-(N-Boc-DOPA), IV. mPEG-NH2 (2.0 g, 0.40 mmoles), N-Boc-L-DOPA dicyclohexylammonium salt (0.80 mmoles), HOBt (1.3 mmoles), and Et3Ν (1.3 mmoles) were dissolved in 20 mL of a 50:50 mixture of DCM and DMF. HBTU (0.80 mmoles) in 10 mL of DCM was then added, and the reaction was carried out under argon at room temperature for 30 minutes. Recovery and purification of the product was performed as described above for I. 1H NMR (500 MHz, CDC13/TMS): δ 6.81-6.60 (m, 3H, C6H3(OH)2-), 6.01 (br, s, IH, OH-), 5.32 (br, s, IH, OH-), 4.22 (br, s, IH, C6H3(OH)2-CH2-CH(N-)-C(O)N-), 3.73-3.38 (m, PEO), 3.07 (m, 2Η, PEO-CH2-NH-C(O)-), 2.73 (t, 2H, C6H3(OH)2-CH2-CH(N-)-C(O)N-), 1.44 (s, 9 H,
(CH3)3C-), 1.25 (s, 3 H, CH3CH2O-). GPC-MALLS: Mw = 6,100, Mw /M„ = 1.02. DOPA Content Determination [00257] The DOPA content of the DOPA-modified PEGs was determined by integration of relevant peaks in the 1H NMR spectrum, and by a colorimetric DOPA assay. In the NMR method, the DOPA content was measured by comparing the integral value of Boc methyl protons at δ = 1.44 to the PEG methylene protons at δ = 3.73-3.38. The DOPA assay was based on the previously described method of Waite and Benedict. Briefly, PEG-DOPA aqueous solutions were treated with nitrite reagent (1.45 M sodium nitrite and 0.41 M sodium molybdate dihydrate) followed by the addition of excess NaOH solution. The absorbance (500 nm) of the mixture was recorded using a Hitachi U-2010 UV/vis spectrophotometer, within 2 to 4 minutes of NaOH addition. A standard curve was constructed using solutions of known DOPA concentration. Formation of PEG-DOPA Hydrogels [00258] To form PEG-DOPA hydrogels, sodium periodate (NaIO4), horseradish peroxidase and hydrogen peroxide (HRP/H2O2), or mushroom tyrosinase and oxygen (MT/O2) were added to solutions of PEG-DOPA (200 mg/mL) in phosphate buffered saline (PBS, pH 7.4). For gelation induced by MT, the PBS was sparged with air for 20 minutes prior to adding MT. Gelation time was qualitatively determined to be when the mixture ceased flowing, as measured by inversion of a vial containing the fluid. Oscillatory Rheometry [00259] Oscillatory rheometry was used to monitor the process of gelation and to determine the mechanic properties of the hydrogels. Cross-linking reagent was added to aqueous solution of PEG-DOPA and the well-mixed solution was loaded onto a Bohlin VOR rheometer. The analysis was performed at a frequency of 0.1 Hz, a strain of 1%, and a 30 mm diameter cone and plate fixture with a cone angle of 2.5°. Spectroscopic Evaluation of DOPA Oxidation [00260] DOPA-modified PEG was dissolved in 10 mM PBS solution (bubbled with argon for HRP/H2O2 and NaIO4 or air for MT experiments). After adding the oxidizing reagent, the time-dependent UV/vis spectra of the solution were monitored at wavelengths from 200 to 700 nm at a scan rate of 800 nm/min. All samples were initially blanked against PBS buffer and recorded at room temperature using a Hitachi U- 2010 UV/vis spectrophotometer. Molecular Weight Analysis [00261] Molecular weights were determined by GPC-MALLS on a DAWN EOS
(Wyatt Technology) using Shodex-OH Pak columns in an aqueous mobile phase (50 mM PBS, 0.1 M NaCl, 0.05% NaN3; pH = 6.0) and a Optilab DSP (Wyatt Technology) refractive index detector. For molecular weight calculations, the experimentally determined dn/dc value of IV (0.136) was used.
Figure imgf000087_0001
II, R = H
Figure imgf000087_0002
III IV
Example 44
Materials and methods Tip modification
[00262] Before the surface modification of silicon nitride (Si3N4) tips, cleaning procedures were performed using O2 plasma (name of a machine) for 3 min and subsequently they were transferred to a piranha solution (sulfuric acid:H2O2 = 8:2) for 30 min. They moved to 20% (v/v) 3-aminopropyltrimethoxysilane in toluene to be functionalized with amines for 30 to 60 min after rinsing with H2O. Two polyethylene glycol (PEG) derivatives were chosen for PEGylation on the AFM tips: mPEG-N- hydroxy succinimide (NHS) (Mw 2000) and Fmoc-PEG-NHS (Mw 3400) (Nektar Inc.). Mixture (Fmoc-PEG-NHS :mPEGNHS = 1 :5-10, 5mM) of PEGs were prepared in 50 mM sodium phosphate buffer, 0.6 M K2SO4, pH 7.8 and chloroform. PEGylation reactions were conducted sequentially first in a sodium phosphate buffer at 40 oC and subsequently in chloroform for 3 hrs in each step. The reason to use PEG mixture is to prevent multiple DOPA binding on TiO2. Fmoc- PEG-NHS provides an amine for Boc- DOPA conjugation after Fmoc cleavage.Piperidine (20% v/v in NMP) was used to deprotect Fmoc for 5 min and subsequently cantilevers were transferred to BOP/HOBt/DOPA (a molar ratio of 1 :1:1, final 8mM in NMP) solution with 10 μL DIPEA. The same procedure was used for Tyrosine modification.
AFM experiment
[00263] All data were collected in AFM instrument (Asylum Research, Santa
Barbara, CA) on top of an inverted Nikon microscope. The spring constant (around 45, 100, and 300 pN/nm by manufacture's information) of an individual cantilever was calibrated by applying the equipartition theorem to the thermal noise spectrum (SI). A drop of water was applied to a pre-cleaned (sonication in organic solvent and O2 plasma) TiO2 surface. Force-distance curves containing PEG elasticity and contour length were selected for further statistical analysis. For the DOPAquinone experiments, all experiments are done in 20 mM Tris, pH 9.8.
Dynamic force experiments
[00264] Loading rate dependent force measurement revealed the energy landscape of DOPA binding (17). A slope (= kBT/xb) of the linear plot (force vs. ln(loading rate)) determines the distance of energy barrier xb along the applied force axis. The binding energy barrier is calculated by the force of logarithmic intercept at zero loading rate from the force transition occurred by pulling rate change and xb from the slope. Silicon nitrite AFM cantilevers (Bio-Levers, Olympus, Japan) were used because of their small string constants (-5 pN/nm and -28 pN/nm). The lowest loading rate of 2 nN/sec in our study was achievable using the pulling rate of 400 nm/sec and the cantilever (-5 pN/nm). The highest loading rate (1500 nN/sec) was produced by 5 μm/sec operation of piezoelectric device and the use of stiff cantilever (300 pN/nm, Veeco). Surface characterization Surfaces were analyzed by X-ray photoelectron microscopy (XPS), (Omicron, Taunusstein Germany) equipped with a nomochromated Al Kα (1486.8 eV) 300 W X-ray source and an electron gun to eliminate charge build-up. Silicon nitride surfaces (0.7 x 0.7 cm2) prepared in the high temperature chamber (ask to Keun Ho) were cleaned and modified as the same procedures described in AFM tip modification. The photoelectron signal from carbon Is orbital was the major indicator for surface modification considering all abundant species of Si, O, and N in Si3N4 surfaces. [00265] While the principles of this invention have been described in connection with specific embodiments, it should be understood clearly that these descriptions are added only by way of examples and are not intended to limit, in any way, the scope of this invention. For instance, the present invention can enhance the adhesive properties of a wide variety of polymeric compositions, whether or not capable of hydrogelation. Likewise, the present invention can be used with various other synthetic techniques well known to those skilled in the art to functionally modify a particular polymeric component for a subsequent coupling and preparation of the corresponding DOPA conjugate. Other advantages, features and benefits will become apparent from the claims filed hereinafter, with the scope thereof as determined by their reasonable equivalents and as would be understood by those skilled in the art.
[00266]

Claims

What is claimed is:
A dihydroxyphenyl (DHPD) adhesive compound of formula (I) wherein
Figure imgf000090_0001
Ri and R2 may be the same or different and are independently selected from the group consisting of hydrogen, saturated and unsaturated, branched and unbranched, substituted and unsubstituted C1- hydrocarbon;
P is separately and independently selected from the group consisting of — NH2, - COOH, -OH, -SH,
Figure imgf000090_0002
wherein Ri and R2 are defined above, a single bond, halogen,
Figure imgf000091_0001
wherein A and A2 are separately and independently selected from the group consisting of H, a single bond, a protecting group, substantially poly(alkyleneoxide) ,
Figure imgf000091_0002
O wherein n ranges between 1 and about 3
Figure imgf000091_0003
C1-6 lower alkyl, or poly(alkylene oxide)-C-C=CH2,
O R3
R3 is defined as above, and D is indicated in Formula (i).
2. A compound according to claim 1 wherein the poly(alkylene oxide) has the structure
Figure imgf000092_0001
wherein R3 and R4 are separately and independently H, or CH3 and m has a value in the range of between 1 and about 250, A is NH2, COOH, OH, -SH, -H, DHPD or a protecting group.
3. A compound according to claim 1 wherein DHPD is of the structure:
Figure imgf000092_0002
4. A compound according to claim 1 wherein DHPD is of the structure:
Figure imgf000093_0001
5. A compound according to claim 1 wherein DHPD is of the structure:
Figure imgf000093_0002
wherein A2 is -OH and Ai is substantially poly(alkylene oxide) of the structure
Figure imgf000093_0003
R3, i and m being defined as in claim 2.
6. A DHPD as in claim 5 wherein the poly(alklene oxide) is a block copolymer of ethylene oxide and propylene oxide.
7. A method of this invention involves adhering substrates to one another comprising the steps of providing DHPD of the structure:
Figure imgf000094_0001
wherein Ri and R2 are defined as above; applying DHPD of the above structure to one or the other or both of the substrates to be adhered; contacting the substrates to be adhered with the DHPD of the above structure therebetween to adhere the substrates to each other;
and optionally repositioning the substrates relative to each other by separating the substrates and recontacting them to each other with the DHPD of the above structure therebetween.
8. A method according to claim 7 wherein Ri and R2 are hydrogen.
9. A method according to claim 7 wherein one or the other of DHPD moieties are of the structure:
Figure imgf000095_0001
10. A method according to claim 7 wherein both DHPD moieties are of the structure:
Figure imgf000095_0002
11. An adhesive comprising a compound of formula (II):
Figure imgf000096_0001
wherein Rls R2, and P are defined as in claim 1.
12. The adhesive of claim 11, wherein the adhesive binds in an aqueous environment.
13. The adhesive of claim 11 , wherein a compound of formula (II) is DOPA.
14. A composition comprising a compound of formula (III)|:
Figure imgf000097_0001
wherein, for each compound of formula (la), Ri and R2 are separately and independently defined as in claim 1 ; and wherein for each compound of formula (la), Pi and P2 are separately and independently defined as P in claim 1, n and m are independently ranging between 0 and about 5, provided that one of m or n is at least 1.
15. A composition according to claim 14 wherein Ri and R2 are all hydrogen.
16. The composition of claim 14, wherein at least one of Pi or P2 comprises substantially poly (alkyleneoxide) .
17. The composition of claim 14, wherein at least one of Pi or P2 comprises at least one site of ethlynic unsaturation.
18. The composition of claim 14, wherein at least 1 of Pi or P2 comprises PEG.
19. The composition of claim 14, wherein at least one of (la) is tri-DOPA, and P is PEG.
20. A composition of claim 14 where Pi and P2 are coupled to each other.
21. A coated medical device comprising a device, and a coating, the coating comprising the composition of claim 14.
22. The coated medical device of claim 20, wherein the device is selected from the group consisting of stents and pacemakers.
23. The coated medical device of claim 14, wherein the coating is biodegradeable.
24. A method of preventing cells or proteins from adhering to a surgical incision site comprising the steps of coating the site with the composition of claim 14.
25. An interaction which is intermediate in strength between a hydrogen bond and a covalent bond, the interaction being substantially reversibly formable, breakable, and reformable.
PCT/US2005/006418 2004-02-27 2005-02-28 Polymeric compositions and related methods of use WO2005118831A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2007500804A JP5133048B2 (en) 2004-02-27 2005-02-28 Polymerized compounds and related methods of use
AU2005250314A AU2005250314A1 (en) 2004-02-27 2005-02-28 Polymeric compositions and related methods of use
CA002557330A CA2557330A1 (en) 2004-02-27 2005-02-28 Polymeric compositions and related methods of use
EP05804747A EP1735456A4 (en) 2004-02-27 2005-02-28 Polymeric compositions and related methods of use

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US54831404P 2004-02-27 2004-02-27
US60/548,314 2004-02-27
US54925904P 2004-03-02 2004-03-02
US60/549,259 2004-03-02

Publications (2)

Publication Number Publication Date
WO2005118831A2 true WO2005118831A2 (en) 2005-12-15
WO2005118831A3 WO2005118831A3 (en) 2007-06-28

Family

ID=35463457

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/006418 WO2005118831A2 (en) 2004-02-27 2005-02-28 Polymeric compositions and related methods of use

Country Status (5)

Country Link
EP (1) EP1735456A4 (en)
JP (1) JP5133048B2 (en)
AU (1) AU2005250314A1 (en)
CA (1) CA2557330A1 (en)
WO (1) WO2005118831A2 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2010501027A (en) * 2006-08-04 2010-01-14 ネリテス コーポレイション Biomimetic compound and synthesis method thereof
WO2010037045A1 (en) 2008-09-28 2010-04-01 Nerites Corporation Multi-armed catechol compound blends
EP2324810A1 (en) * 2009-11-19 2011-05-25 Ivoclar Vivadent AG Filled dental material based on polymerisable dihydroxyphenylalanine derivatives
EP2428503A1 (en) * 2010-09-10 2012-03-14 Justus-Liebig-Universität Gießen Synthesis of tripodal catechol derivatives with a flexible base for functionalising surfaces
US9447407B2 (en) 2011-02-02 2016-09-20 Agency For Science, Technology And Research Double coating procedure for the membranes of bioartificial kidneys
US10177383B2 (en) 2014-03-31 2019-01-08 National Institute For Materials Science Nano-coating material, method for manufacturing same, coating agent, functional material, and method for manufacturing same
WO2021110063A1 (en) * 2019-12-02 2021-06-10 Jiangyin Usun Biochemical Technology Co., Ltd. New conjugates of peptides and polysaccharide

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101046033B1 (en) * 2007-12-06 2011-07-01 주식회사 엘지화학 Highly Adhesive Acrylate Monomer and Its Manufacturing Method
WO2009134303A2 (en) * 2008-03-24 2009-11-05 Clemson University Method and compositions for biofouling deterrence
EP2637707A4 (en) 2010-11-09 2014-10-01 Kensey Nash Corp Adhesive compounds and methods use for hernia repair
JP2012157653A (en) * 2011-02-02 2012-08-23 Agency For Science Technology & Research Double coating method for membrane of bioartificial kidney
JP6041132B2 (en) * 2012-10-19 2016-12-07 国立研究開発法人国立循環器病研究センター Material surface modification method
WO2014118779A1 (en) * 2013-01-31 2014-08-07 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Antifouling materials
CA2906621A1 (en) * 2013-03-15 2014-09-25 Baxter International Inc. Immobilization of active agent on a substrate
WO2014197768A1 (en) 2013-06-07 2014-12-11 Baxter International Inc. Immobilization of an active agent on a substrate using compounds including trihydroxyphenyl groups
WO2015068503A1 (en) * 2013-11-05 2015-05-14 国立大学法人九州工業大学 Polymer, method for producing same, and adhesive composition
EP3305866B1 (en) * 2015-05-26 2020-07-29 Japan Science And Technology Agency Catechol-containing adhesive hydrogel, composition for preparing adhesive hydrogel, and compositions each including said adhesive hydrogel
ES2834994T3 (en) * 2017-08-03 2021-06-21 Fundacio Inst Catala De Nanociencia I Nanotecnologia Catechol derived compounds and their use
KR102369383B1 (en) * 2017-12-07 2022-02-28 주식회사 엘지화학 Carboxylic acid modified nitrile based copolymer latex, method for preparing the copolymer latex, latex composition for dip-forming comprising the copolymer latex and article formed by the composition

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4908404A (en) * 1988-08-22 1990-03-13 Biopolymers, Inc. Synthetic amino acid-and/or peptide-containing graft copolymers
NL1000732C1 (en) * 1995-07-05 1997-01-08 Univ Delft Tech Protein-contg. fraction from mussel feet
US6284267B1 (en) * 1996-08-14 2001-09-04 Nutrimed Biotech Amphiphilic materials and liposome formulations thereof
DE19643007A1 (en) * 1996-10-18 1998-04-23 Martin Dr Schaerfe Bonding agent for improving the adhesion between tooth substance and fillings, bonding agents for artificial teeth, etc
WO2000027802A1 (en) * 1998-11-12 2000-05-18 Ariad Pharmaceuticals, Inc. Bicyclic signal transduction inhibitors, compositions containing them & uses thereof
WO2003008376A2 (en) * 2001-07-20 2003-01-30 Northwestern University Adhesive dopa-containing polymers and related methods of use
WO2006091226A2 (en) * 2004-07-09 2006-08-31 Northwestern University Polymeric compositions and related methods of use

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
E.W. MERRILL, ANN. NYACAD. SCI., vol. 516, 1987, pages 196
METHODS IN ENZYMOLOGY, vol. 107, 1984, pages 397 - 413
OSTUNI ET AL., LANGMUIR, vol. 17, 2001, pages 5605 - 20
PATEL; PRICE, J ORG. CHEM., vol. 30, 1965, pages 3575
See also references of EP1735456A4
SEVER; WILKER, TETRAHEDRON, vol. 57, no. 29, 2001, pages 6139 - 6146

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2010501027A (en) * 2006-08-04 2010-01-14 ネリテス コーポレイション Biomimetic compound and synthesis method thereof
US8916652B2 (en) 2008-09-28 2014-12-23 Kensey Nash Corporation Multi-armed catechol compound blends
EP2348835A1 (en) * 2008-09-28 2011-08-03 KNC NER Acquisition Sub, Inc. Multi-armed catechol compound blends
EP2348835A4 (en) * 2008-09-28 2014-01-22 Knc Ner Acquisition Sub Inc Multi-armed catechol compound blends
WO2010037045A1 (en) 2008-09-28 2010-04-01 Nerites Corporation Multi-armed catechol compound blends
EP2324810A1 (en) * 2009-11-19 2011-05-25 Ivoclar Vivadent AG Filled dental material based on polymerisable dihydroxyphenylalanine derivatives
US8404760B2 (en) 2009-11-19 2013-03-26 Ivoclar Vivadent Ag Filled dental material based on polymerizable dihydroxy-phenylalanine derivatives
EP2428503A1 (en) * 2010-09-10 2012-03-14 Justus-Liebig-Universität Gießen Synthesis of tripodal catechol derivatives with a flexible base for functionalising surfaces
WO2012032085A1 (en) * 2010-09-10 2012-03-15 Justus-Liebig-Universität Giessen Synthesis of trivalent flexible frameworks with ligands comprising catechol units for functionalizing surfaces
US8912332B2 (en) 2010-09-10 2014-12-16 Justus-Liebig-Universitaet Giessen Synthesis of trivalent flexible frameworks with ligands comprising catechol units for functionalizing surfaces
US9447407B2 (en) 2011-02-02 2016-09-20 Agency For Science, Technology And Research Double coating procedure for the membranes of bioartificial kidneys
US10177383B2 (en) 2014-03-31 2019-01-08 National Institute For Materials Science Nano-coating material, method for manufacturing same, coating agent, functional material, and method for manufacturing same
WO2021110063A1 (en) * 2019-12-02 2021-06-10 Jiangyin Usun Biochemical Technology Co., Ltd. New conjugates of peptides and polysaccharide

Also Published As

Publication number Publication date
EP1735456A4 (en) 2011-11-16
WO2005118831A3 (en) 2007-06-28
AU2005250314A1 (en) 2005-12-15
JP2007527871A (en) 2007-10-04
EP1735456A2 (en) 2006-12-27
JP5133048B2 (en) 2013-01-30
CA2557330A1 (en) 2005-12-15

Similar Documents

Publication Publication Date Title
US8815793B2 (en) Polymeric compositions and related methods of use
US7858679B2 (en) Polymeric compositions and related methods of use
JP5133048B2 (en) Polymerized compounds and related methods of use
US7618937B2 (en) Peptidomimetic polymers for antifouling surfaces
US20030087338A1 (en) Adhesive DOPA-containing polymers and related methods of use
Tiu et al. Enhanced adhesion and cohesion of bioinspired dry/wet pressure-sensitive adhesives
Yu et al. Synthetic polypeptide mimics of marine adhesives
AU2007283772B2 (en) Biomimetic compounds and synthetic methods therefor
Tugulu et al. Synthesis of poly (methacrylic acid) brushes via surface-initiated atom transfer radical polymerization of sodium methacrylate and their use as substrates for the mineralization of calcium carbonate
Rodriguez-Emmenegger et al. Quantifying bacterial adhesion on antifouling polymer brushes via single-cell force spectroscopy
Zou et al. It takes walls and knights to defend a castle–synthesis of surface coatings from antimicrobial and antibiofouling polymers
Wang et al. Co-polypeptides of 3, 4-dihydroxyphenylalanine and L-lysine to mimic marine adhesive protein
EP1931718B1 (en) Catechol functionalized polymers and method for preparing them
US10098984B2 (en) Method for grafting polymers on metallic substrates
US20080169059A1 (en) Biomimetic modular adhesive complex: materials, methods and applications therefore
Xu et al. Biomimetic asymmetric polymer brush coatings bearing fencelike conformation exhibit superior protection and antifouling performance
Bai et al. Preparation and characterizations of poly (2-methyl-2-oxazoline) based antifouling coating by thermally induced immobilization
WO2006091226A2 (en) Polymeric compositions and related methods of use
CA2905840A1 (en) Peg-based adhesive phenylic derivatives and methods of synthesis and use
EP3003968A1 (en) Self-assembled micro- and nano-structures
Zhang et al. A facile approach to surface modification on versatile substrates for biological applications
Kohri et al. Adhesion control of branched catecholic polymers by acid stimulation
Lee et al. Biomimetic adhesive polymers based on mussel adhesive proteins
Liu et al. Biomimetic adhesives and coatings based on mussel adhesive proteins
US8907045B2 (en) Biocompatible adhesive polymers

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2557330

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2007500804

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 200580006180.3

Country of ref document: CN

Ref document number: PA/a/2006/009785

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2005804747

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2005250314

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2005250314

Country of ref document: AU

Date of ref document: 20050228

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005250314

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 2005804747

Country of ref document: EP